Skip to main content
Erschienen in: Neuroscience Bulletin 2/2024

Open Access 31.05.2023 | Review

Targeting NAD Metabolism for the Therapy of Age-Related Neurodegenerative Diseases

verfasst von: Feifei Li, Chou Wu, Gelin Wang

Erschienen in: Neuroscience Bulletin | Ausgabe 2/2024

Abstract

As the aging population continues to grow rapidly, age-related diseases are becoming an increasing burden on the healthcare system and a major concern for the well-being of elderly individuals. While aging is an inevitable process for all humans, it can be slowed down and age-related diseases can be treated or alleviated. Nicotinamide adenine dinucleotide (NAD) is a critical coenzyme or cofactor that plays a central role in metabolism and is involved in various cellular processes including the maintenance of metabolic homeostasis, post-translational protein modifications, DNA repair, and immune responses. As individuals age, their NAD levels decline, and this decrease has been suggested to be a contributing factor to the development of numerous age-related diseases, such as cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In pursuit of healthy aging, researchers have investigated approaches to boost or maintain NAD levels. Here, we provide an overview of NAD metabolism and the role of NAD in age-related diseases and summarize recent progress in the development of strategies that target NAD metabolism for the treatment of age-related diseases, particularly neurodegenerative diseases.

Brief Introduction to NAD Metabolism

NAD was first discovered in 1906 by Arthur Harden, who found that boiled yeast extract could stimulate fermentation and alcohol production in vitro [1]. Over the following decades, NAD was purified, its structure was identified, and its function in hydrogen transfer was revealed. The discovery of NAD was the result of the combined efforts of four Nobel Prize winners [2]. For a long time, NAD was regarded as a cofactor in metabolic pathways. However, in recent years, the discovery of NAD-consuming enzymes such as poly (ADP-ribose) polymerases (PARPs), CD38, sirtuins, and SARM1 (sterile alpha and TIR motif containing 1), has revealed the roles of NAD in other important cellular processes, such as the maintenance of genomic stability, protein modification, epigenetic regulation of gene expression, and immune responses. Consequently, NAD is now regarded as the hub of metabolism, and its critical role in aging and disease development is widely appreciated. Below, we outline the biosynthesis and consumption of NAD, as well as its primary functions in a variety of cellular processes.

NAD Biosynthesis

NAD can be synthesized through three distinct pathways (Fig. 1): the de novo synthesis pathway (also known as the kynurenine pathway, or KP for short), the Preiss-Handler pathway, and the salvage pathway. The de novo synthesis of NAD occurs mainly in the liver and begins with dietary tryptophan. Once taken up into cells by SLC6A19, tryptophan is converted into N-formylkynureine by either indoleamine 2,3-dioxygenase (IDO) or tryptophan 2.3-dioxygenase (TDO). N-formylkynureine then undergoes four enzymatic reactions to form α-amino-β-carboxymuconate-ε-semialdehyde (ACMS). Typically, ACMS is converted to picolinic acid in the presence of ACMS decarboxylase. However, ACMS can spontaneously cyclize to form quinolinic acid (QA), which is then condensed by quinolinate phosphoribosyl transferase into nicotinic acid mononucleotide (NaMN) and enters NAD synthesis through the Preiss-Handler pathway. It is important to note that before being condensed into NaMN, QA is a potent neurotoxin with marked free radical-producing properties [3]. The Preiss-Handler pathway was described in 1958 by Jack Preiss and Philip Handler. In this pathway, three enzymes, namely NA phosphoribosyltransferase (NAPRT), NMN adenylyltransferases (NMNATs), and NAD synthase (NADS), sequentially convert the dietary nicotinic acid into NAD as shown in Fig. 1. The salvage pathway is the primary source of NAD biosynthesis in most mammalian cells. Through the salvage pathway, nicotinamide (NAM), the common product of NAD consumption, is converted back into NAD. NAM is catalyzed to NMN by NAMPT, the rate-limiting enzyme in the salvage pathway. NMN is then converted into NAD by NMNATs. NMN can also be generated by nicotinamide riboside kinase (NRK) from dietary nicotinamide riboside (NR).

NAD Consumption

NAD plays two fundamental roles in living organisms. Firstly, through the transition between its oxidized and reduced states, NAD/NADH serves as an electron carrier during glycolysis, oxidative phosphorylation, and fermentation, generating ATP to meet the energy demand of the cell, while providing various intermediate metabolites as building blocks for cell growth and proliferation. During the tricarboxylic acid (TCA) cycle, NAD is converted to NADH, along with the generation of various intermediate metabolites. Subsequently, NADH is converted back to NAD during oxidative phosphorylation, establishing an electron gradient across the inner mitochondrial membrane, which is then used for ATP generation by ATP synthase (complex V) [4, 5]. During this process, the overall levels of NAD and NADH do not change, but metabolic stress can affect the NAD/NADH ratio.
Secondly, NAD plays a crucial role in cellular processes by participating in pathways regulated by NAD-consuming enzymes, including Sirtuins, PARPs, CD38/157, and SARM1 as shown in Fig. 2. Sirtuins (SIRT1-7) are NAD-dependent deacetylases that modify various target proteins, such as histone, peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α, p53, NF-kB, FOXOs, and PARP [6]. They regulate a wide range of processes, including transcription, energy metabolism, circadian rhythm, DNA repair, and inflammation [7, 8]. As the sirtuin cofactor, NAD can have a significant impact on the cellular processes mentioned above, and it appears that NAD's positive role in healthy aging is mostly mediated by elevating sirtuin activity. PARPs are another class of NAD consumer that utilizes NAD as a substrate to catalyze the transfer of the ADP-ribose group from NAD to target proteins, a reaction called PARylation or poly ADP-ribosylation. There are 17 PARP isoforms in humans, PARP1 being the most extensively studied and well-known one [9]. PARP1 plays a critical role in DNA repair and cellular stress responses by catalyzing the PARylation of itself and other proteins after DNA damage caused by UV radiation, reactive oxygen species (ROS), environmental factors, or replication errors. These proteins serve as a scaffold to recruit DNA repair proteins to the site of damage, facilitating the initiation of DNA damage repair mechanisms [9]. The PARP1-mediated PARylation modification is a highly ATP- and NAD-consuming process, and excessive PARP activation results in ATP and NAD depletion, leading to cell death termed parthanatos [10]. CD38 and its paralog CD157 are ectoenzymes that catalyze the breakdown of NAD into NAM and ADP-ribose (ADPR) or cyclic ADP-ribose (cADPR). Initially, CD38 was identified as a surface marker of immune cells, but subsequent research revealed its expression in other cell types, such as endothelial cells and neural cells. Studies have suggested that aging-associated CD38 overexpression is the primary cause of NAD decline [11, 12]. SARM1 is a recently-discovered evolutionarily-conserved NAD hydrolase in the neuronal system [13, 14]. Under normal physiological conditions, the NADase activity of SARM1 is self-inhibited. However, insults that elevate the cellular NMN/NAD ratio can relieve the self-inhibitory status of SARM1 and trigger its NADase activity. This activation causes a significant reduction in neuronal NAD levels, which can lead to metabolic catastrophe and ultimately, neuronal degeneration [15]. The consumption of NAD by NAD-consuming enzymes results in a net decrease in NAD levels, NAM being the common by-product of this process. The NAM is then utilized by the NAD salvage synthesis pathway to regenerate NAD and maintain stable cellular NAD levels. Disruptions of NAD homeostasis have been associated with the development of many diseases, particularly age-related diseases.

The Role of NAD in Aging and Age-Related Neurodegenerative Diseases

Aging is characterized by a progressive loss of physiological integrity, leading to increased susceptibility to a range of diseases, including cancer, diabetes, cardiovascular disorders, and neurodegenerative diseases, which are also known as age-related diseases [16]. The hallmarks of aging include genomic instability, telomere attrition, epigenetic alteration, loss of proteostasis, dysregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intracellular communication [16]. As the hub of metabolism, NAD plays a crucial role in maintaining the health and well-being of organisms ranging from Drosophila to mammals. The decline in NAD levels during aging is widely acknowledged [17]. In this section, we summarize recent progress made in understanding the causes of NAD decline during aging and elucidate how this decline renders organisms susceptible to a variety of age-related diseases, especially neurodegenerative diseases (Fig. 3).

Mechanisms of NAD Decline during Aging

The decline in net NAD levels can result from either decreased synthesis or increased consumption of NAD. According to an isotope-tracing study, circulating NAD precursor levels are largely unchanged and the de novo NAD synthesis from tryptophan is unimpaired during aging. Instead, the primary factor contributing to NAD decline appears to be an accelerated rate of NAD consumption [18]. Consistent with this, another study found that CD38 expression and activity are increased in various tissues, including the liver, adipose tissue, spleen, and skeletal muscles, during aging [19]. CD38-mediated NAD consumption has been suggested to be the main cause of NAD decline during aging. Subsequent studies have revealed that increased inflammation and senescence-associated secretory phenotype (SASP) during aging induce CD38 expression in both endothelial cells and macrophages, leading to increased NAD consumption [11, 20, 21]. Hindered NAMPT expression has also been found in senescent cells, and this further decreases NAD levels by blocking its salvage synthesis [22, 23].

NAD Dysregulation in the Development of Neurodegenerative Diseases

As the hub of metabolism, NAD dysregulation has a significant impact on energy homeostasis, leading to serious age-related metabolic diseases such as diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. Here, we focus on the impact of NAD on neurodegenerative disease. As the most sophisticated and energy-intensive system in the body, the nervous system experiences a decline in NAD levels during normal aging [24] and during the progression of neurodegenerative diseases [25]. The decline in NAD levels occurs even before the onset of neurodegenerative symptoms [26], highlighting its contributory effects on disease progression.
The most well-known age-related neurodegenerative diseases include Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). The common feature of these neurodegenerative diseases is the loss of axons before neurons die [27, 28]. In the typical injury-induced Wallerian degeneration, the loss of NMNAT2, a critical enzyme in the NAD salvage pathway, results in an increased NMN/NAD ratio. This, in turn, activates the NADase activity of SARM1, leading to NAD depletion and eventual axon degeneration [15, 29]. Activation of SARM1 NADase activity has also been reported to play a role in axon loss during neurodegenerative diseases [30]. The expression of Wlds, a chimeric Ube4b/NMNAT1 fusion protein responsible for the “degeneration slow” phenotype after axon injury, has been reported to protect mice from Parkinson’s and Charcot-Marie-Tooth neuropathy [28]. SARM1 knockout has profound neuroprotective effects in several neurodegeneration models, such as diabetic peripheral neuropathy [31], 6-hydroxydopamine-induced loss of dopaminergic axons in the medial forebrain bundle [32], and a mouse model of retinal ganglion cell degeneration [33]. Inhibitors of SARM1 NADase activity are currently under development for the treatment of neurodegenerative diseases (see the section on NAD-boosting strategies).
Dysregulated mitochondrial homeostasis and function resulting from NAD decline are also common in neurodegenerative diseases [34]. As noted earlier, the nervous system is highly energy-demanding and therefore relies heavily on the normal functioning of mitochondria. NAD plays a crucial role, not only in the oxidative phosphorylation function of mitochondria but also in the regulation of mitochondrial homeostasis by increasing mitochondrial-nuclear communication and activating the mitochondrial unfolded protein response [35, 36]. Maintaining normal NAD/NADH levels is critical for the normal integrity and function of mitochondria. In an age-related glaucoma model, Williams et al. found that mitochondrial dysfunction associated with a decline in NAD is one of the first changes within retinal ganglion cells (RGCs) before the onset of disease characteristics [37]. Phenothiazine has been reported to protect the chronic rotenone model of PD by maintaining normal NAD/NADH levels [38].
The relationship between NAD, mitochondria dysfunction, and neurodegeneration was first revealed in xeroderma pigmentosum group A, ataxia-telangiectasia, and Cockayne syndrome, three DNA-repair disorders with severe neurodegeneration [34]. The accumulation of DNA damage in these diseases causes hyperactivation of PARP1, which dramatically consumes NAD. Restricted NAD availability decreases activation of the NAD-SIRT1-PGC-1α axis, impairs mitophagy, and results in the accumulation of damaged mitochondria. NAD supplementation or inhibition of PARP1 activity rescues the mitochondrial phenotype and extends the lifespan [34, 39]. Consistent with this, NAD supplements stimulate mitophagy and accelerate the clearance of damaged mitochondria, as well as the clearance of aggregated amyloid-β and tau protein in AD models [3941]. In another study, decreased alpha-synuclein aggregation by NAD-dependent SIRT3 induction was also reported to increase mitochondrial bioenergetics [42].
The role of mitochondrial dysfunction in neurodegeneration is not well understood. One possibility is that damaged mitochondria have a lower bioenergetic rate, which fails to meet the high energy demand of the neurons. Alternatively, damaged mitochondria may result in more oxidative stress in neurons or both. Oxidative stress is widely recognized as a crucial factor in the onset and progression of neurodegenerative diseases [43]. Environmental toxins that introduce oxidative damage, such as rotenone and paraquat, are also risk factors for the development of neurodegenerative diseases. In a mouse model of Leigh syndrome, which is a severe mitochondrial neurodegenerative disease, continuous exposure to low oxygen levels (11% O2) prevents neurodegeneration and leads to a dramatic extension of the lifespan. This may indicate the contributory effect of oxidative stress in the development of neurodegeneration [44]. Several antioxidants or radical scavenging agents have shown protective effects in preclinical models of neurodegeneration. In a rotenone-induced PD model, the antioxidant compound idebenone decreases lipid peroxidation and mitigates motor neuron impairment [45]. The free-radical scavenger phenothiazine has also been shown to protect against rotenone-induced neuronal toxicity by reducing protein thiol oxidation [38]. 1-Methyl-4-phenylpyridinium (+)-induced blockade of the electron transport chain and reduction in NAD/NADH levels are reversed by mitochondrial uncoupling and the antioxidant agent embelin, which has a neuronal protective effect in the mouse PD model [46]. NAD/NADH itself has been reported to counter oxidative stress derived from the environment and mitochondria as a redox buffer [47]. In addition, elevating NAD levels can decrease ROS levels by improving mitochondrial quality to decrease ROS generation and increase mitochondrial manganese superoxide dismutase function to accelerate ROS elimination [48]. However, the positive effects of several radical-scavenging agents have not been successfully translated into the clinic, possibly because antioxidants cannot reverse established damage to proteins and organelles [49]. Co-treatment with autophagy-inducing strategies, for instance, autophagosome tethering compounds [50], has been suggested to accelerate the clearance of damaged proteins and organelles [49]. Decreased mitochondrial respiratory capacity has also been reported in AD neurons [51]. Nicholls suggested in 2008 that the deleterious consequence of restricting ATP-generating capacity greatly outweighs that of superoxide radicals in intact neurons [52]. Given the highly polarized structure of neuronal cells, damaged mitochondria and insufficient energy generation pose a significant challenge to the long axon terminals. In summary, NAD decline-induced mitochondrial dysfunction can threaten the healthy status of the nervous system by decreasing energy generation and increasing oxidative stress. Whether other mitochondria-related processes, such as the induction of apoptosis or dysregulation of Ca2+ homeostasis, contribute to neurodegeneration requires further study.
Aberrant protein aggregation in the nervous system is another well-known hallmark of neurodegenerative diseases, such as α-synuclein aggregation in PD, amyloid β and Tau aggregation in AD, and TDP (TAR DNA binding protein)-43 aggregation in ALS. For a long time, these aggregated proteins were considered the cause of these neurodegenerative diseases. However, decades of drug development targeting these aggregates have largely failed in the clinic. In recent years, growing evidence suggests that the accumulation of protein aggregates in the nervous system is linked to lysosome dysfunction and the inadequate clearance of misfolded or damaged proteins by autophagy [53]. Neurons are especially vulnerable to protein misfolding and aggregation. Strategies aimed at enhancing the autophagic clearance of aggregated proteins were regarded as the future hope for AD patients. The precise relationship between NAD and autophagy regulation in the nervous system is still largely unknown. Studies conducted in other tissues have shown that sufficient NAD levels are required to maintain lysosomal acidification in heart tissue [54]. A study conducted in breast cancer cells showed that a reduction in SIRT1 levels decreases the expression of the vacuolar-type-ATPase subunit and impairs normal lysosome acidification [55]. In the nervous system, supplementation with the NAD precursor NAM enhances lysosome/autolysosome acidification and reduces autophagosome accumulation in the brains of AD mice [56]. In addition, it has been found that the toxic prion protein induces dramatic NAD depletion, abnormal autophagy activation, and finally neuronal demise [57]. While the exact causal relationship between NAD depletion and abnormal autophagy activation is not yet well established, studies suggest that oxidative stress, which is increased by NAD decline, can accelerate the accumulation of damaged and misfolded proteins, increasing the autophagic stress in the nervous system [49]. Furthermore, the decline in NAD levels has been shown to contribute to the formation of protein aggregation through the nudix homology domain (NHD) that binds NAD. NHD exists in a wide range of proteins and plays a critical role in regulating protein-protein interactions [58]. For example, decreased NAD causes the NAD-binding protein DBC1 to form a complex with PARP1, thus paralyzing its function in DNA damage repair [58]. Aged mice have an increased amount of the DBC1-PARP1 complex, lower PARP1 activity, and increased DNA damage while increasing NAD levels by NMN supplementation can decrease the DBC1-PARP1 complex and reverse all these effects [58, 59]. Thus, a reduction in NAD levels may increase the propensity to form protein complexes through the widely present NHD domain, impairing protein function and increasing autophagic stress at the same time.
Dysregulated cellular processes, including NAD decline, mitochondrial dysfunction, insufficient energy supplementation, redox stress, and decreased autophagy clearance, are the primary drivers of age-related neurodegenerative diseases. These processes can exist prior to the onset of protein aggregates and neurodegenerative symptoms. In general, a decrease in NAD levels disrupts mitochondrial homeostasis, leading to the accumulation of damaged mitochondria. This accumulation can result in inadequate ATP generation and increased ROS generation, which further accelerates NAD decline, mitochondrial damage, DNA damage, and protein aggregation. There is no fixed order of appearance for these events, and all factors are interconnected during the progression of neurodegenerative diseases (as shown in Fig. 3). The appearance of one factor can accelerate the emergence of another, disrupting the homeostatic balance. Maintaining proper NAD levels can increase the ability of the nervous system to counter internal or external disturbance.

Boosting NAD by Increasing NAD Synthesis

Maintaining normal NAD levels is crucial for an organism’s health and homeostasis. Therefore, strategies aimed at boosting or maintaining normal NAD levels are theoretically beneficial. The therapeutic potential of boosting NAD in aging and age-related diseases has been well appreciated [60]. Numerous strategies for boosting NAD have been explored with the hope of curing or delaying neurodegenerative diseases (Table 1).
Table 1:
Summary of strategies for boosting NAD biosynthesis in the treatment of aging and age-related diseases.
Strategy/Targets
Compounds
Diseases
Effects
Ref.
NAD precursor
    
 
NAD
6-OHDA-induced PD model
Direct NAD supplement decreased neuronal damage in a mouse PD model
[61]
 
NAD
Schinzel-Giedion syndrome (SGS)
The neurodegeneration caused by inheritable DNA damage in SGS neurons can be alleviated by NAD supplement
[62]
 
NAD NADPH
Ischemic stroke
The combination of NAD and NADPH provided a greater beneficial effect and a larger therapeutic window in the animal stroke model, by relieving metabolic stress
[63]
 
NAM
glaucoma
NAM supplement protected neurons from mitochondrial and metabolic dysfunctions; decreased the likelihood of glaucoma development by ~10-fold, and protected the optic nerve from excavation and axon loss.
[64, 65]
 
NAM
Mouse 3xTgAD model
NAM treatment for 8 months improved cognitive performance in AD mice and reduced Aβ and pTau accumulation in AD mouse brains.
[56]
 
NAM
Drosophila AD model
NAM supplement rescued mitochondrial defects, protected neurons from Aβ toxicity, and reduced behavioral impairments
[66]
 
NAM
α-synuclein Drosophila PD model
High-dose NAM supplement decreased oxidative stress, increased mitochondrial function, and improved motor function
[67]
 
NMN
Retinal detachment
NMN administration had a neuroprotective effect on photoreceptors after retinal detachment and oxidative stress.
[68]
 
NMN
Retinal ischemia/reperfusion model
NMN supplement significantly suppressed retinal functional damage and inflammation.
[69]
 
NMN
ischemia
NMN dramatically protected the hippocampal CA1 from ischemic injury and showed strong protective effects against ischemic brain injury.
[70]
 
NR
3xTgAD/polβ(+/-) mice
NR improved the cognitive function of AD mice and decreased brain DNA damage, neuroinflammation, and apoptosis while increasing SIRT3 activity in AD brains.
[71]
 
NR
Tg2576 mouse AD model
NR treatment enhanced PGC-1α expression, β-secretase degradation in the brain, and improved cognitive function and synaptic plasticity.
[72]
 
NR
APP/PS1 mouse AD model
NR increased AD brain NAD levels, decreased neuronal inflammation, induced mitophagy, and improved cognitive and synaptic function
[73]
 
NR
Mouse aging
NR delayed neuronal stem cell senescence and increased mouse lifespan
[74]
 
NR
NMDA-induced brain damage
Intracortical administration of NR reduced brain damage induced by NMDA injection. And delayed NMDA-induced axon degeneration in cultured neurons.
[75]
 
NR
Acute/chronic RGC damage
NR enhanced RGC survival in both optic nerves crush-induced acute RGC damage and ocular hypertension-induced chronic RGC damage.
[76]
 
NR
ALS
NR supplement delayed motor neuron degeneration and decreased neuroinflammation in hSOD1-linked ALS mouse model.
[77]
 
NR
ALS
NR activated the mitochondrial unfolded protein response and improved neurogenesis in adult ALS mouse brains.
[78]
 
NRC
ischemia
NRC increased energy supply and promoted cognitive function recovery after mouse brain ischemia
[79]
NAMPT activators
    
 
P7C3-A20
Traumatic brain injury model (TBI)
P7C3-A20 was neuroprotective and promoted endogenous reparative strategies after TBI.
[80]
 
P7C3-S243
TBI
P7C3-S243 blocked axon degeneration and preserved normal synaptic activity, learning, and memory in TBI mice
[81]
 
P7C3-S243
P7C3-A20
6-OHDA model of PD
Compounds blocked dopaminergic neuron death and preserved normal motor behavior.
[82]
 
P7C3
Sciatic nerve crush injury model
P7C3 treatment doubled the neuron survival period after injury and promoted axon regeneration.
[83]
 
P7C3-A20
Paclitaxel-induced -peripheral neuropathy
P7C3-A20 treatment provided robust neuroprotection towards paclitaxel-induced peripheral neuropathy
[84]
 
P7C3-A20
Ischemic stroke model
P7C3-A20 administration significantly improved stroke-induced damage even when taken 6 hours after ischemia
[85]
 
NAT, NAT-5R
Paclitaxel-induced peripheral neuropathy
NAT and NAT-5R alleviated paclitaxel-induced peripheral neuropathy in mice.
[86]
NADH dehydrogenase modulators
    
 
β-lapachone
Age-related hearing loss, Cisplatin-induced hearing loss
β-lapachone improved age-related hearing loss and cisplatin- induced hearing loss by increasing NAD levels through modulation of NQO1 activity
[87, 88]
Natural products in boosting NAD
    
 
Apigenin
LPS-induced neurotoxicity
Apigenin protected mice from LPS-induced neurotoxicity and cognitive decline, which may act by modulating NAD/NADH levels and boosting of SIRT3 activity.
[89, 90]
 
Fisetin
6-OHDA-treated SH-SY5Y cells
Fisetin protected neuronal cells from 6-OHDA-induced apoptotic cell death by modulating the PI3K-Akt pathway. Fisetin was reported to be a PARP1 inhibitor and SIRT1 activator
[9193]
 
Embelin
PD
Embelin protected MPP(+)-induced N27 dopaminergic cell apoptosis and showed a protective effect in the MPTP mouse model of PD
[46]
The levels of NAD in an organism are maintained through a balance between its synthesis and consumption. Strategies aimed at boosting NAD levels can be categorized into two classes: increasing its synthesis or blocking its consumption. In this section, we summarize recent progress in strategies aimed at accelerating NAD synthesis. In the next section, we discuss strategies for slowing down NAD consumption. As shown in Fig. 1, dietary tryptophan, nicotinic acid, and NAM are the starting materials for NAD synthesis. Therefore, it is reasonable to consider supplementing these precursors to boost NAD production. In addition, the inadequate function of the NAD salvage pathway is also an important cause of NAD decline [78]. Therefore, manipulating the salvage pathway activity or supplementing NAD precursors has been widely studied as a promising NAD-boosting strategies.

NAD Precursors as Neuroprotective Agents

Several NAD precursors have been used both in vitro and in vivo to enhance NAD levels. Here, we discuss the efficacy and safety of these NAD precursors. Direct supplementation of NAD has been found to be effective in reducing neuronal damage by alleviating mitochondrial dysfunction in a mouse model of PD [61]. Moreover, direct supplementation of NAD shows neuroprotective effects in Schinzel-Giedion syndrome, a type of juvenile neurodegenerative disease [62]. In multiple ischemic stroke models, the combination of NAD with a small dose of NADPH has a more potent neuroprotective effect by increasing ATP levels and decreasing ROS levels [63]. However, in an NMDA-induced excitotoxicity-related axon degeneration model, direct supplementation of NAD was found to have a weaker protective effect than NR supplementation [75].
The de novo synthesis of NAD from tryptophan is a significant means by which cells produce NAD. As illustrated in Fig. 1, the de novo synthesis of NAD is a complex process that involves the participation of many enzymes and the generation of various intermediates. Dysregulation of tryptophan metabolism has been implicated in various neurodegenerative diseases. In biomarker-related studies, patients with neurodegenerative diseases have been found to exhibit an imbalance in tryptophan metabolism [94]. Deletion of TDO has been shown to enhance neurogenesis in the hippocampus and subventricular zone of the mouse brain [95], and inhibition of the KP pathway enzymes, such as IDO and TDO, have also been reported to be neuroprotective [96, 97]. Inhibiting the activity of the KP pathway enzyme TDO has also been reported to extend lifespan [98]. It is worth noting that the majority of the neuroprotective effects resulting from KP pathway manipulations are in contrast to its NAD synthesizing property. As the majority of NAD is produced through the salvage pathway, the relationship between diseases and NAD generation from the kynurenine pathway is not well established [99, 100]. A study on NAD synthetic fluxes suggested that the liver synthesizes NAD from tryptophan, releasing NAM into the circulatory system to be used by other tissues [101]. Further investigation is necessary to determine the impact of the KP pathway on NAD metabolism in the context of aging and disease. Nicotinic acid and NAM, the other two dietary NAD precursors, known as vitamin B3, are well known for their ability to cure severe pellagra. The incidence of pellagra decrease as the quality of life improves. Nonetheless, nicotinic acid has been found to possess other pharmacological activities at higher concentrations, such as treating dyslipidemia and age-related neurological disorders, including AD, PD, and Huntington’s disease [102]. In a recent study, treatment with nicotinic acid for 12 months increased muscle strength and mitochondrial biogenesis in 5 patients with progressive external ophthalmoplegia (PEO), which is a type of mitochondrial myopathy [103]. However, there have been fewer studies on the use of nicotinic acid, possibly due to the lack of efficacy demonstrated in two long-term clinical trials [104, 105] and the undesirable side-effects associated with its use [102, 105]
NAM serves as the starting material for the salvage synthesis of NAD, and its supplementation has been reported to have a protective effect in glaucoma [64, 65]. It can also protect against degeneration and alleviate symptoms in a Drosophila model of AD [66], as well as decrease oxidative stress, and improve motor function in a Drosophila PD model [67]. In the 3xTgAD mouse model, 8 months of NAM supplementation was shown to enhance cognitive performance in AD mice by reducing toxic protein aggregation in brain tissue [56]
The NAD-elevating potential of NMN and its relevant protective effects on neuronal system diseases have also been extensively studied [48, 68, 106]. The protective effects of NMN supplementation against retinal detachment-induced photoreceptor degeneration [68], ischemia/reperfusion-induced retinal damage [69], and ischemia-induced hippocampal CA1 injury [70] have all been confirmed. Mechanistic studies have revealed that the function of NMN involves the prevention of age-related gene expression, enhancement of mitochondrial oxidative metabolism, maintenance of mitochondrial-nuclear communication, the elevation of SIRT1 protein expression/activity, and the SIRT3-dependent global decrease in mitochondrial protein acetylation [68, 107110].
The safety of administering oral NMN or NMN derivatives has been evaluated both preclinically and clinically. In a preclinical rodent model, administration of 500 mg/kg/day of NMN for 91 days showed no adverse events, while the highest dose (2000 mg/kg/day) led to reduced body weight and diminished food consumption in rats, effects similar to those seen with the reference item nicotinamide riboside chloride (NRC) dosed at 1740 mg/kg/day [111]. In mice, administration of 300 mg/kg/day NMN for 14 days also showed no acute toxicity [112]. In a clinical trial, a microcrystalline unique polymorph β-NMN formula called MIB-626 was found to be well-tolerated when taken once or twice daily at a dose of 1000 mg for 14 consecutive days by overweight or obese adults [113]. A promising effect of NMN has also been shown in preclinical and clinical models of aging, as NMN administration for 40 days by pre-aging mice (16 months) or human volunteers (45–60 years old) significantly increased telomere length in peripheral blood mononuclear cells [114]. In a long-term study in mice, 12 months of NMN administration not only proved to be safe but also suppressed the age-related increase in body weight, enhanced energy metabolism, showed a positive effect on physical activity, and increased insulin sensitivity when compared to normal chow-fed mice [109].
However, the use of NMN to elevate NAD levels has been questioned. Firstly, there is an ongoing debate about whether NMN is cell permeable [115, 116]. Originally, it was believed that NMN was metabolized to NR extracellularly and then taken up by cells [115]. NR was then catalyzed to NMN inside cells by NRK to enter the NAD salvage synthesis pathways [117]. In 2019, the first NMN transporter Slc12a8 was identified [116], but debate concerning the existence and function of slc12a8 continues [115]. Secondly, concerns about the safety of NMN administration have been raised due to recent findings that an increase in NMN/NAD ratio leads to the activation of SARM1, which is undesirable [15]. Despite these concerns, the known beneficial effects of NMN supplements suggest that there may be unknown mechanisms involved in NMN uptake and metabolism that require future investigation.
NR is another NAD precursor that has been extensively studied. Research has shown that, during normal aging, NR can delay neural stem cell senescence and increase the lifespan of mice [74]. A recent phase I clinical trial on PD patients found that oral NR supplementation is safe and results in mild clinical improvement [118]. In several preclinical studies, NR showed protective effects against various nervous system diseases or damage, including AD [7173], NMDA-induced brain damage [75], retinal ganglion cell damage [76], and an hSOD1-linked ALS mouse disease [77]. In a mouse brain ischemia model, the NR salt NRC was found to increase the energy supply and promote cognitive function recovery [79]. Studies also suggest that, in most AD models, NR exerts its function by accelerating the clearance of protein aggregation, inducing mitophagy, and improving mitochondrial function in neurons [73, 119]. NR supplementation can also activate the mitochondrial unfolded protein response and improve neurogenesis in adult ALS mouse brains [78].
Despite promising results, concerns have been raised about the stability of NR in the circulatory system, and its use for NAD production is limited by the rate-limiting NRK. To overcome these obstacles, Giroud et al. reported that NRH, the reduced form of NR, is a more potent and faster-acting NAD precursor than NR, and can prevent cisplatin-induced acute kidney injury when taken orally [120]. The in vivo effects of NAD or other NAD precursors in aging or age-related diseases have also been summarized elsewhere [121, 122].

Development of NAMPT Activators

The NAD salvage pathway serves as the primary source of cellular NAD. As previously noted, NAMPT and NMNATs are enzymes involved in this salvage pathway, and activating these two enzymes is expected to boost cellular NAD levels. Researchers at the University of Texas Southwestern Medical Center reported the discovery of the first NAMPT activator, P7C3 [123]. They found that P7C3 and its analog, P7C3-A20, enhance the activity of purified NAMPT in vitro, increase NAD levels within cells, protect axons after injury, and have protective effects in mouse models of PD, ALS, traumatic brain injury, paclitaxel-induced peripheral neuropathy, neonatal nerve injury, and developmental delays associated with Down syndrome, a genetic disorder causing intellectual disability [8085, 123125]. The neuroprotective effects of P7C3 in rodents have been successfully translated to primates [126], and the P7C3 treatment remains effective even when administered one year after traumatic brain injury [127]. The NAD-boosting activity of P7C3 has also been reported to be beneficial in age-related diabetic heart and skeletal muscle [128, 129]. By enhancing NAD salvage pathway activity, P7C3 and its analogs exert a positive impact on mitochondrial function and the overall metabolism of cells, resulting in the reported benefits.
The success of P7C3 instills confidence in the development of NAMPT activators as neuroprotective strategies. Following P7C3 and its analog, another NAMPT activator called SBI-797812 has been reported [130]. This compound activates NAMPT-mediated NMN production in a concentration-dependent manner in the presence of ATP, with an EC50 value of 0.37 ± 0.06 μmol/L. However, in the absence of ATP, SBI-797812 slightly inhibits the NMN synthesis activity of NAMPT. Further biochemical analysis has revealed that SBI-797812 induces a significant shift in the reaction equilibrium towards the NAMPT forward reaction. In addition, SBI-797812 relieves the NAD-mediated NAMPT inhibition, thus allowing for boosting NAD levels. Functional validation has confirmed that SBI-797812 increases cellular NMN and NAD levels in cultured cells. An in vivo study found a 1.3-fold increase in NAD in the liver [130]. However, this compound is still undergoing optimization [131, 132], and its impact on the nervous system has not been reported yet.
Our recent study screened 50,000 compounds and identified another NAMPT activator, NAT [86], which binds to NAMPT at a 1:1 ratio with a binding constant of ~500 nmol/L. NAT can significantly increase the catalytic activity (Vmax) of NAMPT for its substrates NAM and phosphoribosyl diphosphate (PRPP). Co-crystal structural analysis has revealed that NAT sits at one end of the active channel of the NAMPT homodimer and slightly affects the binding of NAMPT to its substrates. Structural optimization has generated a more potent NAT analog, NAT-5r [133]. A functional study confirmed that NAT and NAT-5r promote NAD salvage pathway activity and induce metabolic reprogramming by increasing glycolysis, the TCA cycle, and fatty acid oxidation processes inside cells. NAT and NAT-5r have been found to be capable of protecting mice from paclitaxel-induced peripheral neuropathy and promoting the proliferation and self-renewal of primary neural stem cells when cultured in vitro [86]. The potential use of NAT and NAT-5r in age-related diseases is under investigation.
In addition to developing small-molecular activators, the expression level of NAMPT can also be regulated, and its activity can be regulated through post-translational modification. For example, the NAD-dependent deacetylase SIRT6 has been reported to upregulate NAMPT activity by directly deacetylating NAMPT [134]. In a kidney ischemia/reperfusion model, downregulation of the NAMPT protein levels induced by reperfusion can be prevented by ERK1/2 inhibition through a small-molecular MEK1/2 inhibitor, trametinib [135]. Furthermore, triterpenes extracted from Panax notoginseng leaf are capable of protecting against oxygen-glucose deprivation/re-oxygenation-induced ischemia injury in mice and SH-SY5Y cells by up-regulating NAMPT expression [136].

Manipulation of NMNAT Activity

NMNAT is the enzyme that catalyzes the second step of the NAD salvage synthesis, and normal levels and function of NMNAT are critical in maintaining NAD levels. Mammals have three NMNAT isoforms that are localized in distinct cellular compartments. Specifically, NMNAT1 is found in the nucleus, NMNAT2 is found in the Golgi and Golgi-derived vesicles, and NMNAT3 is found in the mitochondria. Among them, NMNAT2 has the shortest half-life and plays a critical role in preserving the integrity of neuronal axons [137, 138]. Reduced levels of NMNAT2 protein have been reported in various conditions, including aged oocytes [139], the brain tissue of individuals with AD [140, 141], and heart tissue affected by cardiac hypertrophy [142]. In each of these cases, exogenous overexpression of NMNAT2 has been found to have beneficial effects. Overexpression of NMNAT1, which is the nuclear form of NAD synthase, has a protective effect in hTau mice [143], and gene therapy such as AAV-mediated NMNAT2 overexpression shows promising results against glaucomatous neurodegeneration [144]. Studies involving the overexpression of NMNATs have consistently shown positive results, confirming the beneficial effect of increasing the activity of these enzymes. However, there have been no reports of small molecules capable of enhancing the activity of NMNAT2 or slowing its degradation. Haubrich et al. developed a high-throughput screening assay to identify NMNAT modulators and found several NMNAT1 inhibitors from a pool of 912 compounds. Among them, 2,3-dibromo-1,4-naphthoquinone was identified as the most potent inhibitor [145]. Although no activators were identified in this screening, it is important to note that the library of 912 compounds is relatively small, and conducting the same screening scheme on a larger library could potentially yield exciting results.

Generating NAD through NADH Dehydrogenase

NADH dehydrogenases, including NAD(P)H: Quinone Oxidoreductase 1 (NQO1) and cytochrome b5 reductase 3, catalyze the conversion of NADH to NAD. Mice that overexpress these two enzymes exhibit phenotypes similar to those seen with calorie restriction, including increased lifespan, improved physical performance, and decreased chronic inflammation [146]. The natural products β-lapachone and dunnione promote the conversion of NADH to NAD by increasing NQO1 function [88, 147]. Increasing NAD levels with β-lapachone has been found to effectively prevent age-related and cisplatin-induced hearing loss [87, 88]. Modulating NQO1 activity to increase NAD levels also provides protection against chemotherapy-induced nephrotoxicity, small intestinal damage, cardiac dysfunction, and lung fibrosis [147150]. Overexpression of another NADH dehydrogenase, the NADH-ubiquinone oxidoreductase (NDI1), has been shown to ameliorate the loss of optical nerve axons and retinal ganglion cells in a mouse model of experimental autoimmune encephalomyelitis, protecting mice from permanent visual loss [151, 152], even though the impact of NDI1 expression on NAD levels was not mentioned.

Natural Products in Neuroprotection

There is a large body of research focused on the therapeutic potential of natural products for the treatment of neurodegenerative diseases. Although the specific targets of these natural products remain unclear, their reported effects include ameliorating oxidative stress through activation of the Nrf2 system, up-regulating NQO1 and heme oxygenase-1 levels, and increasing NAD/NADH levels. Considering the focus of our paper, we only mention a few relevant studies here. Apigenin, a flavonoid found in nature, has been found to be capable of inhibiting CD38 activity and maintaining NAD/NADH levels [89]. Apigenin has been shown to protect mice from LPS-induced neurotoxicity and cognitive impairment by promoting mitochondrial fusion and mitophagy. Its mechanism of action involves maintaining NAD/NADH levels and boosting SIRT3 activity [90]. Fisetin is another plant flavonoid that has been shown to protect against 6-OHDA-induced neuronal cell death through activation of the PI3K-Akt pathway [91]. The neuroprotective effects of fisetin might also be exerted through the inhibition of PARP1 activity [92] and activation of the SIRT1 activity [93]. Another natural product, embelin, has been found to be capable of increasing NAD/NADH levels and enhancing SIRT1-PGC1α activity by its mitochondrial uncoupling effects. These properties of embelin have been found to be beneficial against MPTP-induced PD [46]. The traditional Chinese herbal prescription Dihuang-Yinzi has long been used to treat neurodegenerative diseases such as AD. Investigation into its mechanism of action revealed that the ability to increase NAD content and enhance energy metabolism may contribute to its neuroprotective activity [153].

Maintenance of NAD Levels by Modulating NAD-Consuming Enzymes

Reducing unnecessary consumption of NAD represents another promising approach for elevating NAD levels. As noted above, enzymes that consume NAD include sirtuins, PARPs, CD38/157, and SARM1. In the following sections, we discuss strategies targeting each of these enzymes individually (Table 2).
Table 2:
Strategies by modulating NAD-consuming enzymes.
Targets
Compounds
Disease model
Effects
Ref.
Sirtuin activity modulators
    
SIRT1 activator
Resveratrol
Cerebral ischemia (CI)
SIRT1 activation modulates neuronal survival in aged CI mice in an Akt-dependent manner
[154]
SIRT1 activator
Sulfonamide derivatives
6-OHDA-treated neuronal cell
SIRT1 activation by sulfonamide derivatives can protect SH-SY5Y from 6-OHDA-induced cell death
[155]
SIRT1 activator
NeuroHeal
Peripheral nerve axotomy
NeuroHeal can activate the pro-survival autophagy process and protect pups from peripheral nerve axotomy by the concomitant activation of SIRT1 and the PI3K/Akt pathway
[156]
SIRT2 inhibitor
AGK2
Ischemic stroke model
SIRT2 inhibitor AGK2 or sirt2 knockout have neuroprotective effects in the transient middle cerebral artery occlusion mouse model
[157]
SIRT2 inhibitor
Thioamide 53
 
Thioamide 53 can promote neurite outgrowth of Neuro-2a cells
[158]
PARP inhibitors
    
 
Olaparib
Schinzel-Giedion syndrome (SGS)
The neurodegeneration caused by inheritable DNA damage can be alleviated by PARP1 inhibition
[62]
 
10e
PD
10e protects neurons from α-synuclein pre-formed fiber-mediated neurotoxicity and helps maintain normal NAD levels
[159]
 
PJ34
Rat forebrain ischemia
PJ34 almost completely inhibits neuroinflammation and reduces CA1 neuronal death by 84%.
[160]
 
PJ34
Rotenone-treated Drosophila
PARP1 inhibition by PJ34 reduces dSARM expression, and protects Drosophila from rotenone-induced loss of locomotor ability.
[161]
 
INO1001
Aortic cross-clamping-induced ischemia/reperfusion
INO1001 markedly protects the spinal cord from aortic occlusion-induced injury.
[162]
 
INO1001
Brain traumatic injury
INO1001 significantly reduces microglial activation and increases neuronal survival after TBI.
[163]
 
INO1001
R6/2 mutant Huntington’s disease mouse model
INO1001 prolongs R6/2 mutant mouse survival, and reduces severe signs of neurological dysfunction compared to vehicle control
[164]
SARM1 inhibitors
    
 
Berberine Chloride
Chronic acrylamide-induced axon destruction
Berberine treatment significantly ameliorates axonal degeneration, alleviates pathological changes in the sciatic nerve and spinal cord, and improves neurobehavioral symptoms in acrylamide-treated rats.
[165]
 
DSRM-3716
Rotenone-induced axon degeneration
SARM1 inhibition by DSRM-3716 rescues rotenone-treated axons that are fated to degenerate
[166]
 
NB-3
Nerve injury model
Vincristine-induced neuropathy model
NB-3 is covalently conjugated with SARM1 product ADPR and exerts a potent protective effect against nerve injury
[167]
 
EV-99
Vacor and vincristine-induced neuropathy
EV-99 covalently binds to C311 of SARM1, and protects axons from vincristine- and vacor-induced neurite degeneration in cultured dorsal root ganglion neurons
[168]
CD38 inhibitors
    
 
78c
age
78c can reverse age-related NAD decline, and improve age-related physiological and metabolic parameters
[169]
 
78c
age
Increases lifespan and health span of naturally aged mice
[170]
 
apigenin
Diabetes, cell senescence, neuroinflammation
Apigenin has protective effects in age-related diabetes, age-related cell senescence, and LPS-induced neuroinflammation and cognitive impairment
[89, 171173]
 
MK-0159
Ischemia/reperfusion
MK-0159 shows strong protection against I/R-induced myocardial damage
[174]
Other enzymes
    
Phosphodiesterase (PDE)10A
papaverine
Quinolinic acid-treated primary cortical neuron
Papaverine can increase cellular NAD levels, restore mitochondrial membrane potential, reduce ROS levels, and show neuroprotective activity
[175]
PDE4
Roflumilast
Quinolinic acid-treated primary cortical neuron
Roflumilast can increase intracellular NAD content and protects primary cortical neurons from quinolinic acid-induced toxicity
[176]
PED4
Roflupram
MPP(+)-treated neuronal cells
Roflupram protects dopaminergic neurons from MPP(+)-induced apoptotic cell death via CREB/PGC1α pathways
[177]
 
Roflupram
Rotenone-treated SH-SY5Y cells
Roflupram can increase the NAD/NADH levels, activate lysosome function, and reduce α-syn levels in rotenone-treated neuronal cells.
[178]

Modulation of Sirtuin Activity

Sirtuins are a class of protein deacetylases that are conserved through evolution and play a critical role in the regulation of metabolism. There are 7 isoforms of sirtuins in humans, known as SIRT1-7, with different cellular localization and functions [179]. The impact of different sirtuin isoforms on aging and age-related diseases is controversial, and whether inhibiting or activating sirtuin activity depends on the specific isoform involved and requires careful consideration on a disease-by-disease basis [180, 181].
Increasing SIRT1 activity has been shown to be neuroprotective, and strategies to elevate SIRT1 protein levels or boost SIRT1 activity are considered potential interventions for aging and neurodegenerative diseases. Elevating SIRT1 protein levels using various methods, including the small molecule A03, AAV-mediated SIRT1 overexpression, and induced SIRT1 expression, have been shown to have protective effects in an AD mouse model [182], N2a cells transfected with amyloid precursor protein [183], RGC neurons [184], and high-fat diet-induced diabetic neuropathy [185]. In a mouse model of cerebral ischemia, activation of SIRT1 was found to exert a neuroprotective effect by activating the JNK/ERK/MAPK/Akt pathway in aged mice [154]. Small-molecule activators of SIRT1, sulfonamide, and its derivatives, were found to protect against 6-OHDA-induced SH-SY5Y cell death [155]. NeuroHeal, an AI-designed compound to target neurodegenerative diseases, has been shown to activate both the SIRT1 and the PI3K/Akt signaling pathways, which converge on FOXO3 deacetylation and phosphorylation. This activation, in turn, leads to the induction of a pro-survival autophagy process that protects rat pups from peripheral nerve axotomy [156]. Knocking out sirt3 in mice also shows significant neuroprotection against ischemia/reperfusion-induced damage or stroke injury. However, this effect has been ultimately attributed to a compensatory rise in the SIRT1 protein levels, rather than the absence of SIRT3 itself [186].
The impact of SIRT2 on aging and age-related diseases remains unclear and controversial, highlighting the need for further research in this area [187]. SIRT2 activity is required for the maintenance of hematopoietic stem cells, oligodendrocyte fate decision, and neuron remyelination during aging [188190]. Moreover, it has been reported that SIRT2 in oligodendrocytes can be transported into axons via exosomes to help neurons maintain ATP homeostasis in axons [191]. However, SIRT2 activity has also been shown to be neurotoxic [182, 192]. Promising protective effects have been reported in a mouse model of ischemic stroke when either sirt2 is knocked out or its activity is inhibited using the SIRT2-specific inhibitor AGK2 [157]. The reported protective effects may occur through multiple mechanisms, including downregulation of the FOXO3 and MAPK signaling pathways [192], as well as blockade of necrosis, which requires SIRT2-dependent RIP1/3 deacetylation [193]. Thioamide 53, another SIRT2-selective inhibitor, has been found to promote neurite outgrowth in Neuro-2a cells while inhibiting the growth of breast cancer cells [158]. Further evaluation is needed to assess the neuroprotective effect of thioamide 53.
As NAD-dependent protein deacetylases, sirtuins have a wide range of substrates and their functions are quite complex. It is reasonable to expect that sirtuins may have complicated effects under different physiological and pathological conditions. Therefore, the development of highly selective inhibitors or activators for different sirtuin isoforms is required, and their use under different conditions must be carefully considered. Basic research aimed at elucidating the physiological roles of sirtuin proteins will be crucial for the successful development and application of sirtuin modulators in the clinic. In addition, studies on the development of different sirtuin modulators have been reviewed elsewhere [180].

Modulation of PARP Activity

PARP is another essential NAD consumer, and its overactivation can lead to the depletion of cellular NAD and ATP. Initially, PARP inhibitors were developed for treating BRCA1/2-deficient breast and ovarian cancers due to the synthetic lethality relationship between BRCA1/2 and PARP [194]. Gradually, the potential of PARP inhibitors in treating aging and neuroprotective diseases has also been appreciated. Studies demonstrated that deleting PARP can boost NAD levels, which may have a beneficial impact on AD. Furthermore, in human patients, certain PARP polymorphisms have been associated with reduced risk and severity of AD [66]. PARP inhibition by Olaparib or 10e was found to have neuroprotective effects in a mouse model of transient cerebral ischemia [195] and in in vitro neuronal models of PD [159]. Purines, such as hypoxanthine, inosine, and adenosine, can reduce PARP activity [196]. The well-known PARP inhibitor INO1001 has been found to reduce spinal cord injury in an ischemia/reperfusion model [162], protect the brain from traumatic injury [163], and has a neuroprotective effect in the R6/2 mouse model of Huntington’s disease [164]. Another PARP1 inhibitor, PJ34, was reported to suppress neuroinflammation and increase neuronal cell survival in a rat model of forebrain ischemia/reperfusion [160]. PJ34 can also protect Drosophila from rotenone-induced locomotor disability by maintaining NAD levels and inhibiting dSARM expression [161].

Modulation of SARM1 Activity

SARM1 has been identified as a critical mediator of axon degeneration in the nervous system following traumatic injury, as well as in various neurodegenerative diseases. Developing SARM1 inhibitors was assumed to protect the neuronal system from degeneration and was widely viewed as a promising neuroprotective strategy [197]. Deletion of the sarm1 gene in mice was shown to protect against several types of axon degeneration, including diabetic neuropathy [31], traumatic brain injury [198], and vincristine-induced neuropathy [199], giving confidence in the development of small-molecular SARM1 inhibitors. High-throughput screening identified berberine chloride and zinc chloride as the first noncompetitive SARM1 inhibitors with modest potency [200]. The neuroprotective potential of berberine chloride against chronic acrylamide-induced axon destruction was confirmed in a recent study [165]. Subsequently, a more potent and selective isoquinoline inhibitor, DSRM-3716, was discovered and found able to replicate the sarm1 knockout phenotype and protect axons from axotomy-induced or mitochondrial dysfunction-induced degeneration [166]. Additional investigation into the mechanism of action of DSRM-3716 (designated as 1) revealed that a base exchange reaction happens between 1 and the NAM moiety of NAD, leading to the formation of a new compound, 1AD, which is a bona fide SARM1 orthostatic inhibitor [197]. Cryo-EM structural studies revealed that the allosteric activator NMN binds to the ARM domain, leading to a significant reorientation of the ARM domain and the formation of a two-stranded TIR domain, as well as an orthosteric site. 1AD binds to the orthosteric site spanning two TIR domain molecules and directly blocks the NADase activity [197]. Bratkowski and colleagues described a new class of SARM1 inhibitors that can intercept NAD hydrolysis by covalently conjugating with ADPR to form small-molecule ADPR adducts, which can effectively inhibit SARM1 activity. The neuroprotective effect of one such inhibitor, NB-3, has been demonstrated in preclinical nerve injury models and a vincristine-induced neuropathy model [167]. Considering that SARM1 is activated by a high NMN/NAD ratio, Sasaki et al. proposed that the combination of the NAMPT inhibitor FK866 with nicotinic acid riboside can be used to decrease the NMN level and increase the NAD precursor NaMN level, thereby helping to maintain a low NMN/NAD ratio and effectively blocking SARM1 activation. NaMN has also been reported to be capable of binding to the allosteric pocket of SARM1 and helping maintain the auto-inhibited configuration of the SARM1 ARM domain [201]. The authors suggested that the allosteric binding pocket of SARM1 enables the development of potent SARM1 inhibitors for the treatment of neurodegenerative disorders. Another study by Feldman reported that the tryptoline acrylamide EV-99 can specifically and covalently bind to C311 of the ARM domain, and protect the axon from vacor- or vincristine-induced axon degeneration [168]. Since the NMN/NAD ratio mediates the self-inhibition status of SARM1 [13], agents that can increase NAD levels may have the potential to inhibit SARM1 activity. For instance, Sarkar recently reported that the PARP inhibitor PJ34 can inhibit rotenone-induced SARM1 activation [161].

Modulation of CD38 Activity

CD38 is an important cell surface ectoenzyme widely expressed in various types of cells and functions as another NAD hydrolase [202, 203]. Reports have shown that age-related upregulation of CD38 is the main cause of NAD decline during aging [19]. Therefore, developing CD38 inhibitors could be a useful strategy for maintaining NAD levels in the aging process. CD38 knockout has been found to decrease AD pathology in an APP/PS mouse model [204] and protects mice against ischemic brain damage [205]. However, CD38 knockout does not affect ALS mouse survival in a hSOD1-linked ALS mouse model [77], indicating that CD38-mediated NAD consumption may not be involved in hSOD1-induced ALS. Tarrago et al. reported a potent and specific CD38 inhibitor, 78c, which can reverse the age-related NAD decline and improve age-related physiological and metabolic parameters [169]. Moreover, 78c has been found to be capable of increasing the lifespan and health span of naturally aged mice, improving exercise performance, endurance, and metabolic function [170]. Another study suggested that 78c exerts its activity by forming an adduct with the CD38 product ADPR, and the 78c-ADPR adduct formed acts as a potent CD38 inhibitor [167]. Apigenin, a flavonoid natural product, is capable of inhibiting CD38 activity [89], and its ability to maintain NAD/NADH levels has been confirmed in several studies [89, 171, 172]. The protective role of apigenin has mainly been reported in age-related metabolic disorders such as diabetes [89, 172], age-related cell senescence [173], LPS-induced neuroinflammation, neurotoxicity, and cognitive impairment [90, 171]. Recently, another potent CD38 inhibitor, MK-0159, has been reported. Its IC50 against CD38 enzymatic activity in vitro is 3 nmol/L [174]. MK-0159 has been found to exert stronger protection against I/R injury-induced myocardial damage than NR or 78c, making it an attractive candidate for various CD38 overexpression-related diseases and conditions.

Other Enzyme Inhibitors

PAP has been shown to increase cellular NAD levels and exert neuronal protective activity by inhibiting phosphodiesterase-10A [175]. Similarly, the phosphodiesterase-4 (PDE4) inhibitor, Roflumilast, has been reported to increase the intracellular NAD content and has a protective effect against quinolinic acid-induced human primary cortical neuron toxicity [176]. A Roflumilast analog, Roflupram, has also been shown to exert a neuroprotective effect by increasing the cellular NAD/NADH levels and activating lysosomal function [178]. It has been suggested that the neuroprotective effect of PDE4 inhibitors may be attributed to the activation of the CREB/PGC1α pathway and the resulting improvement of metabolic parameters [177]. Nicotinamide N-methyltransferase (NNMT) is an enzyme associated with an impaired NAD salvage pathway and has been found in aged muscles. Inhibitors of NNMT have been reported to increase myoblast regeneration and functional recovery after skeletal muscle injury [206].

Conclusions

As our understanding of aging expands, the pursuit of healthy aging has become an increasingly desirable goal. However, despite this aspiration, healthy aging remains out of reach for most people. To address this issue, it is crucial that we continue to make new scientific discoveries in order to gain a systematic understanding of the aging process and develop innovative strategies to prevent, delay, or even cure age-related diseases. Furthermore, it is imperative that we effectively translate this research into clinical practice in order to benefit human beings. It is important to note that aging is a systematic and progressive process, and developing one age-related disease can increase the risk of developing additional age-related conditions. As a result, older individuals often suffer from multiple age-related diseases concurrently. Unfortunately, the current medical care system typically treats patients in a disease-specific manner, which can be inefficient. To address this issue, we propose a comprehensive approach to treating age-related diseases, with a focus on boosting NAD levels as an ideal choice.
The decline in NAD levels during aging and age-related diseases has been identified as a targetable process for promoting healthy aging and/or curing or delaying the progression of age-related diseases. Several strategies for boosting NAD levels have been proposed, and we summarized recent advances above. Currently, there are three categories of strategies under development for increasing NAD levels: providing NAD precursors, enhancing NAD synthesis pathway activity, and inhibiting NAD-consuming enzyme activity. It is currently unclear whether any of these strategies is superior to the others. While NAD precursor supplements are the most frequently studied strategy, it is important to note that this approach may not be effective or even harmful if NAD synthesis pathways are blocked in certain diseases. For example, NMNAT2, which is critical for NAD salvage synthesis, has been reported to be downregulated in several neurodegenerative diseases. Therefore, manipulating the activity or protein levels of NAD synthesis enzymes is a second crucial strategy for maintaining a healthy dynamic status of NAD. However, one potential concern with this strategy is the variability in the half-life of NAD across different tissues, which can range from 15 minutes to 15 hours [101]. As the physiological importance of this difference is still unknown, it is important to consider the tissue selectivity of this approach in case the disruption of this difference may result in potential adverse effects. Modulating the activity of NAD-consuming enzymes is the third avenue for increasing NAD levels. Small molecules or antibodies targeting CD38 provide promising activity, as increased CD38 activity has been found to be responsible for the global decline in NAD levels during aging. SARM1 inhibitors are under extensive development now, as activation of SARM1 results in axon degeneration. However, modulators of sirtuins and PARPs need to be developed with caution, as the functions of these two NAD consumers are complicated and sometimes controversial. Many beneficial effects of NAD are exerted by sirtuins, and normal PARP activity is critical in maintaining genomic stability. Therefore, the physiological and pathological functions of different NAD consumers should be carefully considered during the development of their modulators.
Boosting NAD as a potential therapy for age-related neurodegenerative diseases is still in an early stage of development. A better understanding of NAD metabolism and regulation is necessary and requires further basic research. Investigating the roles of various NAD-consuming enzymes in NAD degradation across different tissues and subcellular compartments is crucial. While promising preclinical results exist, there is currently limited evidence of therapeutic benefits in clinical populations. Therefore, it is crucial to continue efforts to successfully translate these findings into clinical practice. In addition, it is still unknown whether one NAD precursor is superior to another, and systematic comparisons are needed. It is also essential to determine whether the use of different NAD precursors should be tailored to specific diseases and stages of the disease.
When developing NAD-boosting strategies, three key points must always be kept in mind. Firstly, since aging and age-related diseases are chronic and require long-term interventions, safety is a vital concern when developing compounds or strategies. A careful examination of the tradeoff between benefits and risk factors is necessary. Secondly, determining a safety range for NAD boosting is essential since excessive levels of NAD can be harmful, as shown by reported oxidative cytotoxicity induced by the potent NAD precursor NRH [207]. Lastly, since NAD is in a dynamic equilibrium between synthesis and consumption, it is perhaps more important to keep NAD in a healthy salvaging state rather than elevating the stable state of NAD levels. With these considerations in mind, safe and efficient NAD-boosting strategies can pave the way for a future of healthy aging.

Acknowledgements

We apologize for omitting related studies and citations due to space limits. This review was supported by the National Natural Science Foundation of China (91949101 and 81872874), the Tsinghua-Peking Center for Life Sciences, the Beijing Advanced Innovation Center for Structural Biology, the Tsinghua-Toyota Joint Research Fund, and the Tsinghua University Initiative Scientific Research Program.

Conflict of interest

The authors declare that there are no conflicts of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Harden A, Young WJ. The alcoholic ferment of yeast-juice. Proc R Soc Lond B 1906: 405–420. Harden A, Young WJ. The alcoholic ferment of yeast-juice. Proc R Soc Lond B 1906: 405–420.
2.
Zurück zum Zitat Berger F, Ramírez-Hernández MH, Ziegler M. The new life of a centenarian: Signalling functions of NAD(P). Trends Biochem Sci 2004, 29: 111–118.PubMedCrossRef Berger F, Ramírez-Hernández MH, Ziegler M. The new life of a centenarian: Signalling functions of NAD(P). Trends Biochem Sci 2004, 29: 111–118.PubMedCrossRef
3.
Zurück zum Zitat Sas K, Robotka H, Toldi J, Vécsei L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J Neurol Sci 2007, 257: 221–239.PubMedCrossRef Sas K, Robotka H, Toldi J, Vécsei L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J Neurol Sci 2007, 257: 221–239.PubMedCrossRef
4.
6.
Zurück zum Zitat Kane AE, Sinclair DA. Sirtuins and NAD+ in the development and treatment of metabolic and cardiovascular diseases. Circ Res 2018, 123: 868–885.PubMedPubMedCentralCrossRef Kane AE, Sinclair DA. Sirtuins and NAD+ in the development and treatment of metabolic and cardiovascular diseases. Circ Res 2018, 123: 868–885.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab 2014, 25: 138–145.PubMedCrossRef Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab 2014, 25: 138–145.PubMedCrossRef
9.
Zurück zum Zitat Spiegel JO, van Houten B, Durrant JD. PARP1: Structural insights and pharmacological targets for inhibition. DNA Repair (Amst) 2021, 103: 103125.PubMedCrossRef Spiegel JO, van Houten B, Durrant JD. PARP1: Structural insights and pharmacological targets for inhibition. DNA Repair (Amst) 2021, 103: 103125.PubMedCrossRef
10.
Zurück zum Zitat Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 2011, 4: ra20.PubMedPubMedCentralCrossRef Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 2011, 4: ra20.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Chini CCS, Peclat TR, Warner GM, Kashyap S, Espindola-Netto JM, de Oliveira GC, et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD+ and NMN levels. Nat Metab 2020, 2: 1284–1304.PubMedPubMedCentralCrossRef Chini CCS, Peclat TR, Warner GM, Kashyap S, Espindola-Netto JM, de Oliveira GC, et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD+ and NMN levels. Nat Metab 2020, 2: 1284–1304.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Wu S, Zhang R. CD38-expressing macrophages drive age-related NAD+ decline. Nat Metab 2020, 2: 1186–1187.PubMedCrossRef Wu S, Zhang R. CD38-expressing macrophages drive age-related NAD+ decline. Nat Metab 2020, 2: 1186–1187.PubMedCrossRef
13.
Zurück zum Zitat Jiang Y, Liu T, Lee CH, Chang Q, Yang J, Zhang Z. The NAD+-mediated self-inhibition mechanism of pro-neurodegenerative SARM1. Nature 2020, 588: 658–663.ADSPubMedCrossRef Jiang Y, Liu T, Lee CH, Chang Q, Yang J, Zhang Z. The NAD+-mediated self-inhibition mechanism of pro-neurodegenerative SARM1. Nature 2020, 588: 658–663.ADSPubMedCrossRef
14.
Zurück zum Zitat Essuman K, Summers DW, Sasaki Y, Mao X, DiAntonio A, Milbrandt J. The SARM1 toll/interleukin-1 receptor domain possesses intrinsic NAD+ cleavage activity that promotes pathological axonal degeneration. Neuron 2017, 93: 1334–1343.e5.PubMedPubMedCentralCrossRef Essuman K, Summers DW, Sasaki Y, Mao X, DiAntonio A, Milbrandt J. The SARM1 toll/interleukin-1 receptor domain possesses intrinsic NAD+ cleavage activity that promotes pathological axonal degeneration. Neuron 2017, 93: 1334–1343.e5.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Figley MD, Gu W, Nanson JD, Shi Y, Sasaki Y, Cunnea K, et al. SARM1 is a metabolic sensor activated by an increased NMN/NAD+ ratio to trigger axon degeneration. Neuron 2021, 109: 1118–1136.e11.PubMedPubMedCentralCrossRef Figley MD, Gu W, Nanson JD, Shi Y, Sasaki Y, Cunnea K, et al. SARM1 is a metabolic sensor activated by an increased NMN/NAD+ ratio to trigger axon degeneration. Neuron 2021, 109: 1118–1136.e11.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Yoshino J, Baur JA, Imai SI. NAD+ intermediates: The biology and therapeutic potential of NMN and NR. Cell Metab 2018, 27: 513–528.PubMedCrossRef Yoshino J, Baur JA, Imai SI. NAD+ intermediates: The biology and therapeutic potential of NMN and NR. Cell Metab 2018, 27: 513–528.PubMedCrossRef
18.
Zurück zum Zitat McReynolds MR, Chellappa K, Chiles E, Jankowski C, Shen Y, Chen L, et al. NAD+ flux is maintained in aged mice despite lower tissue concentrations. Cell Syst 2021, 12: 1160–1172.e4.PubMedPubMedCentralCrossRef McReynolds MR, Chellappa K, Chiles E, Jankowski C, Shen Y, Chen L, et al. NAD+ flux is maintained in aged mice despite lower tissue concentrations. Cell Syst 2021, 12: 1160–1172.e4.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Camacho-Pereira J, Tarragó MG, Chini CCS, Nin V, Escande C, Warner GM, et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab 2016, 23: 1127–1139.PubMedPubMedCentralCrossRef Camacho-Pereira J, Tarragó MG, Chini CCS, Nin V, Escande C, Warner GM, et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab 2016, 23: 1127–1139.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Chini C, Hogan KA, Warner GM, Tarragó MG, Peclat TR, Tchkonia T, et al. The NADase CD38 is induced by factors secreted from senescent cells providing a potential link between senescence and age-related cellular NAD+ decline. Biochem Biophys Res Commun 2019, 513: 486–493.PubMedPubMedCentralCrossRef Chini C, Hogan KA, Warner GM, Tarragó MG, Peclat TR, Tchkonia T, et al. The NADase CD38 is induced by factors secreted from senescent cells providing a potential link between senescence and age-related cellular NAD+ decline. Biochem Biophys Res Commun 2019, 513: 486–493.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, et al. Senescent cells promote tissue NAD+ decline during ageing via the activation of CD38+ macrophages. Nat Metab 2020, 2: 1265–1283.PubMedPubMedCentralCrossRef Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, et al. Senescent cells promote tissue NAD+ decline during ageing via the activation of CD38+ macrophages. Nat Metab 2020, 2: 1265–1283.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Pi C, Ma C, Wang H, Sun H, Yu X, Gao X, et al. MiR-34a suppression targets Nampt to ameliorate bone marrow mesenchymal stem cell senescence by regulating NAD+-Sirt1 pathway. Stem Cell Res Ther 2021, 12: 271.PubMedPubMedCentralCrossRef Pi C, Ma C, Wang H, Sun H, Yu X, Gao X, et al. MiR-34a suppression targets Nampt to ameliorate bone marrow mesenchymal stem cell senescence by regulating NAD+-Sirt1 pathway. Stem Cell Res Ther 2021, 12: 271.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Gong H, Chen H, Xiao P, Huang N, Han X, Zhang J, et al. MiR-146a impedes the anti-aging effect of AMPK via NAMPT suppression and NAD+/SIRT inactivation. Signal Transduct Target Ther 2022, 7: 66.PubMedPubMedCentralCrossRef Gong H, Chen H, Xiao P, Huang N, Han X, Zhang J, et al. MiR-146a impedes the anti-aging effect of AMPK via NAMPT suppression and NAD+/SIRT inactivation. Signal Transduct Target Ther 2022, 7: 66.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci U S A 2015, 112: 2876–2881.ADSPubMedPubMedCentralCrossRef Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci U S A 2015, 112: 2876–2881.ADSPubMedPubMedCentralCrossRef
25.
Zurück zum Zitat van der Velpen V, Rosenberg N, Maillard V, Teav T, Chatton JY, Gallart-Ayala H, et al. Sex-specific alterations in NAD+ metabolism in 3xTg Alzheimer’s disease mouse brain assessed by quantitative targeted LC-MS. J Neurochem 2021, 159: 378–388.PubMedPubMedCentralCrossRef van der Velpen V, Rosenberg N, Maillard V, Teav T, Chatton JY, Gallart-Ayala H, et al. Sex-specific alterations in NAD+ metabolism in 3xTg Alzheimer’s disease mouse brain assessed by quantitative targeted LC-MS. J Neurochem 2021, 159: 378–388.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Bertrand M, Decoville M, Meudal H, Birman S, Landon C. Metabolomic nuclear magnetic resonance studies at presymptomatic and symptomatic stages of Huntington’s disease on a Drosophila model. J Proteome Res 2020, 19: 4034–4045.PubMedCrossRef Bertrand M, Decoville M, Meudal H, Birman S, Landon C. Metabolomic nuclear magnetic resonance studies at presymptomatic and symptomatic stages of Huntington’s disease on a Drosophila model. J Proteome Res 2020, 19: 4034–4045.PubMedCrossRef
27.
Zurück zum Zitat Loreto A, Angeletti C, Gu W, Osborne A, Nieuwenhuis B, Gilley J, et al. Neurotoxin-mediated potent activation of the axon degeneration regulator SARM1. Elife 2021, 10: e72823.PubMedPubMedCentralCrossRef Loreto A, Angeletti C, Gu W, Osborne A, Nieuwenhuis B, Gilley J, et al. Neurotoxin-mediated potent activation of the axon degeneration regulator SARM1. Elife 2021, 10: e72823.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Conforti L, Gilley J, Coleman MP. Wallerian degeneration: An emerging axon death pathway linking injury and disease. Nat Rev Neurosci 2014, 15: 394–409.PubMedCrossRef Conforti L, Gilley J, Coleman MP. Wallerian degeneration: An emerging axon death pathway linking injury and disease. Nat Rev Neurosci 2014, 15: 394–409.PubMedCrossRef
30.
Zurück zum Zitat Figley MD, DiAntonio A. The SARM1 axon degeneration pathway: Control of the NAD+ metabolome regulates axon survival in health and disease. Curr Opin Neurobiol 2020, 63: 59–66.PubMedPubMedCentralCrossRef Figley MD, DiAntonio A. The SARM1 axon degeneration pathway: Control of the NAD+ metabolome regulates axon survival in health and disease. Curr Opin Neurobiol 2020, 63: 59–66.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Cheng Y, Liu J, Luan Y, Liu Z, Lai H, Zhong W, et al. Sarm1 gene deficiency attenuates diabetic peripheral neuropathy in mice. Diabetes 2019, 68: 2120–2130.PubMedPubMedCentralCrossRef Cheng Y, Liu J, Luan Y, Liu Z, Lai H, Zhong W, et al. Sarm1 gene deficiency attenuates diabetic peripheral neuropathy in mice. Diabetes 2019, 68: 2120–2130.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Peters OM, Weiss A, Metterville J, Song L, Logan R, Smith GA, et al. Genetic diversity of axon degenerative mechanisms in models of Parkinson’s disease. Neurobiol Dis 2021, 155: 105368.PubMedPubMedCentralCrossRef Peters OM, Weiss A, Metterville J, Song L, Logan R, Smith GA, et al. Genetic diversity of axon degenerative mechanisms in models of Parkinson’s disease. Neurobiol Dis 2021, 155: 105368.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Finnegan LK, Chadderton N, Kenna PF, Palfi A, Carty M, Bowie AG, et al. SARM1 ablation is protective and preserves spatial vision in an in vivo mouse model of retinal ganglion cell degeneration. Int J Mol Sci 2022, 23: 1606.PubMedPubMedCentralCrossRef Finnegan LK, Chadderton N, Kenna PF, Palfi A, Carty M, Bowie AG, et al. SARM1 ablation is protective and preserves spatial vision in an in vivo mouse model of retinal ganglion cell degeneration. Int J Mol Sci 2022, 23: 1606.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Fang EF, Scheibye-Knudsen M, Brace LE, Kassahun H, SenGupta T, Nilsen H, et al. Defective mitophagy in XPA via PARP-1 hyperactivation and NAD(+)/SIRT1 reduction. Cell 2014, 157: 882–896.PubMedPubMedCentralCrossRef Fang EF, Scheibye-Knudsen M, Brace LE, Kassahun H, SenGupta T, Nilsen H, et al. Defective mitophagy in XPA via PARP-1 hyperactivation and NAD(+)/SIRT1 reduction. Cell 2014, 157: 882–896.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD(+)/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 2013, 154: 430–441.PubMedPubMedCentralCrossRef Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD(+)/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 2013, 154: 430–441.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Gomes AP, Price NL, Ling AJY, Moslehi JJ, Montgomery MK, Rajman L, et al. Declining NAD(+) induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 2013, 155: 1624–1638.PubMedPubMedCentralCrossRef Gomes AP, Price NL, Ling AJY, Moslehi JJ, Montgomery MK, Rajman L, et al. Declining NAD(+) induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 2013, 155: 1624–1638.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Williams PA, Harder JM, Foxworth NE, Cochran KE, Philip VM, Porciatti V, et al. Vitamin B3 modulates mitochondrial vulnerability and prevents glaucoma in aged mice. Science 2017, 355: 756–760.ADSPubMedPubMedCentralCrossRef Williams PA, Harder JM, Foxworth NE, Cochran KE, Philip VM, Porciatti V, et al. Vitamin B3 modulates mitochondrial vulnerability and prevents glaucoma in aged mice. Science 2017, 355: 756–760.ADSPubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Tapias V, McCoy JL, Greenamyre JT. Phenothiazine normalizes the NADH/NAD+ ratio, maintains mitochondrial integrity and protects the nigrostriatal dopamine system in a chronic rotenone model of Parkinson’s disease. Redox Biol 2019, 24: 101164.PubMedPubMedCentralCrossRef Tapias V, McCoy JL, Greenamyre JT. Phenothiazine normalizes the NADH/NAD+ ratio, maintains mitochondrial integrity and protects the nigrostriatal dopamine system in a chronic rotenone model of Parkinson’s disease. Redox Biol 2019, 24: 101164.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Yang B, Dan X, Hou Y, Lee JH, Wechter N, Krishnamurthy S, et al. NAD+ supplementation prevents STING-induced senescence in Ataxia telangiectasia by improving mitophagy. Aging Cell 2021, 20: e13329.PubMedPubMedCentralCrossRef Yang B, Dan X, Hou Y, Lee JH, Wechter N, Krishnamurthy S, et al. NAD+ supplementation prevents STING-induced senescence in Ataxia telangiectasia by improving mitophagy. Aging Cell 2021, 20: e13329.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci 2019, 22: 401–412.PubMedPubMedCentralCrossRef Fang EF, Hou Y, Palikaras K, Adriaanse BA, Kerr JS, Yang B, et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci 2019, 22: 401–412.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Sorrentino V, Romani M, Mouchiroud L, Beck JS, Zhang H, D’Amico D, et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature 2017, 552: 187–193.ADSPubMedPubMedCentralCrossRef Sorrentino V, Romani M, Mouchiroud L, Beck JS, Zhang H, D’Amico D, et al. Enhancing mitochondrial proteostasis reduces amyloid-β proteotoxicity. Nature 2017, 552: 187–193.ADSPubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Park JH, Burgess JD, Faroqi AH, DeMeo NN, Fiesel FC, Springer W, et al. Alpha-synuclein-induced mitochondrial dysfunction is mediated via a sirtuin 3-dependent pathway. Mol Neurodegener 2020, 15: 5.PubMedPubMedCentralCrossRef Park JH, Burgess JD, Faroqi AH, DeMeo NN, Fiesel FC, Springer W, et al. Alpha-synuclein-induced mitochondrial dysfunction is mediated via a sirtuin 3-dependent pathway. Mol Neurodegener 2020, 15: 5.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Bhat AH, Dar KB, Anees S, Zargar MA, Masood A, Sofi MA, et al. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed Pharmacother 2015, 74: 101–110.PubMedCrossRef Bhat AH, Dar KB, Anees S, Zargar MA, Masood A, Sofi MA, et al. Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomed Pharmacother 2015, 74: 101–110.PubMedCrossRef
44.
Zurück zum Zitat Grange RMH, Sharma R, Shah H, Reinstadler B, Goldberger O, Cooper MK, et al. Hypoxia ameliorates brain hyperoxia and NAD+ deficiency in a murine model of Leigh syndrome. Mol Genet Metab 2021, 133: 83–93.PubMedPubMedCentralCrossRef Grange RMH, Sharma R, Shah H, Reinstadler B, Goldberger O, Cooper MK, et al. Hypoxia ameliorates brain hyperoxia and NAD+ deficiency in a murine model of Leigh syndrome. Mol Genet Metab 2021, 133: 83–93.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Avcı B, Günaydın C, Güvenç T, Yavuz CK, Kuruca N, Bilge SS. Idebenone ameliorates rotenone-induced parkinson’s disease in rats through decreasing lipid peroxidation. Neurochem Res 2021, 46: 513–522.PubMedCrossRef Avcı B, Günaydın C, Güvenç T, Yavuz CK, Kuruca N, Bilge SS. Idebenone ameliorates rotenone-induced parkinson’s disease in rats through decreasing lipid peroxidation. Neurochem Res 2021, 46: 513–522.PubMedCrossRef
46.
Zurück zum Zitat Rao SP, Sharma N, Kalivendi SV. Embelin averts MPTP-induced dysfunction in mitochondrial bioenergetics and biogenesis via activation of SIRT1. Biochim Biophys Acta BBA Bioenerg 2020, 1861: 148157.CrossRef Rao SP, Sharma N, Kalivendi SV. Embelin averts MPTP-induced dysfunction in mitochondrial bioenergetics and biogenesis via activation of SIRT1. Biochim Biophys Acta BBA Bioenerg 2020, 1861: 148157.CrossRef
47.
Zurück zum Zitat Ghosh D, Levault KR, Brewer GJ. Relative importance of redox buffers GSH and NAD(P)H in age-related neurodegeneration and Alzheimer disease-like mouse neurons. Aging Cell 2014, 13: 631–640.PubMedPubMedCentralCrossRef Ghosh D, Levault KR, Brewer GJ. Relative importance of redox buffers GSH and NAD(P)H in age-related neurodegeneration and Alzheimer disease-like mouse neurons. Aging Cell 2014, 13: 631–640.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Klimova N, Long A, Kristian T. Nicotinamide mononucleotide alters mitochondrial dynamics by SIRT3-dependent mechanism in male mice. J Neurosci Res 2019, 97: 975–990.PubMedPubMedCentralCrossRef Klimova N, Long A, Kristian T. Nicotinamide mononucleotide alters mitochondrial dynamics by SIRT3-dependent mechanism in male mice. J Neurosci Res 2019, 97: 975–990.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Giordano S, Darley-Usmar V, Zhang J. Autophagy as an essential cellular antioxidant pathway in neurodegenerative disease. Redox Biol 2014, 2: 82–90.PubMedCrossRef Giordano S, Darley-Usmar V, Zhang J. Autophagy as an essential cellular antioxidant pathway in neurodegenerative disease. Redox Biol 2014, 2: 82–90.PubMedCrossRef
50.
Zurück zum Zitat Zhang H, An P, Fei Y, Lu B. Modeling the degradation effects of autophagosome tethering compounds. Neurosci Bull 2021, 37: 255–260.PubMedCrossRef Zhang H, An P, Fei Y, Lu B. Modeling the degradation effects of autophagosome tethering compounds. Neurosci Bull 2021, 37: 255–260.PubMedCrossRef
51.
Zurück zum Zitat Theurey P, Connolly NMC, Fortunati I, Basso E, Lauwen S, Ferrante C, et al. Systems biology identifies preserved integrity but impaired metabolism of mitochondria due to a glycolytic defect in Alzheimer’s disease neurons. Aging Cell 2019, 18: e12924.PubMedPubMedCentralCrossRef Theurey P, Connolly NMC, Fortunati I, Basso E, Lauwen S, Ferrante C, et al. Systems biology identifies preserved integrity but impaired metabolism of mitochondria due to a glycolytic defect in Alzheimer’s disease neurons. Aging Cell 2019, 18: e12924.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Nicholls DG. Oxidative stress and energy crises in neuronal dysfunction. Ann N Y Acad Sci 2008, 1147: 53–60.ADSPubMedCrossRef Nicholls DG. Oxidative stress and energy crises in neuronal dysfunction. Ann N Y Acad Sci 2008, 1147: 53–60.ADSPubMedCrossRef
53.
Zurück zum Zitat Lee JH, Yang DS, Goulbourne CN, Im E, Stavrides P, Pensalfini A, et al. Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci 2022, 25: 688–701.PubMedPubMedCentralCrossRef Lee JH, Yang DS, Goulbourne CN, Im E, Stavrides P, Pensalfini A, et al. Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci 2022, 25: 688–701.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Yagi M, Toshima T, Amamoto R, Do Y, Hirai H, Setoyama D, et al. Mitochondrial translation deficiency impairs NAD+-mediated lysosomal acidification. EMBO J 2021, 40: e105268.PubMedPubMedCentralCrossRef Yagi M, Toshima T, Amamoto R, Do Y, Hirai H, Setoyama D, et al. Mitochondrial translation deficiency impairs NAD+-mediated lysosomal acidification. EMBO J 2021, 40: e105268.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, et al. Loss of sirtuin 1 alters the secretome of breast cancer cells by impairing lysosomal integrity. Dev Cell 2019, 49: 393-408.e7.PubMedPubMedCentralCrossRef Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, et al. Loss of sirtuin 1 alters the secretome of breast cancer cells by impairing lysosomal integrity. Dev Cell 2019, 49: 393-408.e7.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Liu D, Pitta M, Jiang H, Lee JH, Zhang G, Chen X, et al. Nicotinamide forestalls pathology and cognitive decline in Alzheimer mice: Evidence for improved neuronal bioenergetics and autophagy procession. Neurobiol Aging 2013, 34: 1564–1580.PubMedCrossRef Liu D, Pitta M, Jiang H, Lee JH, Zhang G, Chen X, et al. Nicotinamide forestalls pathology and cognitive decline in Alzheimer mice: Evidence for improved neuronal bioenergetics and autophagy procession. Neurobiol Aging 2013, 34: 1564–1580.PubMedCrossRef
57.
Zurück zum Zitat Zhou M, Ottenberg G, Sferrazza GF, Hubbs C, Fallahi M, Rumbaugh G, et al. Neuronal death induced by misfolded prion protein is due to NAD+ depletion and can be relieved in vitro and in vivo by NAD+ replenishment. Brain 2015, 138: 992–1008.PubMedPubMedCentralCrossRef Zhou M, Ottenberg G, Sferrazza GF, Hubbs C, Fallahi M, Rumbaugh G, et al. Neuronal death induced by misfolded prion protein is due to NAD+ depletion and can be relieved in vitro and in vivo by NAD+ replenishment. Brain 2015, 138: 992–1008.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Li J, Bonkowski MS, Moniot S, Zhang D, Hubbard BP, Ling AJ, et al. A conserved NAD+ binding pocket that regulates protein-protein interactions during aging. Science 2017, 355: 1312–1317.ADSPubMedPubMedCentralCrossRef Li J, Bonkowski MS, Moniot S, Zhang D, Hubbard BP, Ling AJ, et al. A conserved NAD+ binding pocket that regulates protein-protein interactions during aging. Science 2017, 355: 1312–1317.ADSPubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Mendelsohn AR, Larrick JW. The NAD+/PARP1/SIRT1 axis in aging. Rejuvenation Res 2017, 20: 244–247.PubMedCrossRef Mendelsohn AR, Larrick JW. The NAD+/PARP1/SIRT1 axis in aging. Rejuvenation Res 2017, 20: 244–247.PubMedCrossRef
60.
61.
Zurück zum Zitat Shan C, Gong YL, Zhuang QQ, Hou YF, Wang SM, Zhu Q, et al. Protective effects of β- nicotinamide adenine dinucleotide against motor deficits and dopaminergic neuronal damage in a mouse model of Parkinson’s disease. Prog Neuro Psychopharmacol Biol Psychiatry 2019, 94: 109670.CrossRef Shan C, Gong YL, Zhuang QQ, Hou YF, Wang SM, Zhu Q, et al. Protective effects of β- nicotinamide adenine dinucleotide against motor deficits and dopaminergic neuronal damage in a mouse model of Parkinson’s disease. Prog Neuro Psychopharmacol Biol Psychiatry 2019, 94: 109670.CrossRef
62.
Zurück zum Zitat Banfi F, Rubio A, Zaghi M, Massimino L, Fagnocchi G, Bellini E, et al. SETBP1 accumulation induces P53 inhibition and genotoxic stress in neural progenitors underlying neurodegeneration in Schinzel-Giedion syndrome. Nat Commun 2021, 12: 4050.ADSPubMedPubMedCentralCrossRef Banfi F, Rubio A, Zaghi M, Massimino L, Fagnocchi G, Bellini E, et al. SETBP1 accumulation induces P53 inhibition and genotoxic stress in neural progenitors underlying neurodegeneration in Schinzel-Giedion syndrome. Nat Commun 2021, 12: 4050.ADSPubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Huang Q, Sun M, Li M, Zhang D, Han F, Wu JC, et al. Combination of NAD+ and NADPH offers greater neuroprotection in ischemic stroke models by relieving metabolic stress. Mol Neurobiol 2018, 55: 6063–6075.PubMedCrossRef Huang Q, Sun M, Li M, Zhang D, Han F, Wu JC, et al. Combination of NAD+ and NADPH offers greater neuroprotection in ischemic stroke models by relieving metabolic stress. Mol Neurobiol 2018, 55: 6063–6075.PubMedCrossRef
64.
Zurück zum Zitat Williams PA, Harder JM, Cardozo BH, Foxworth NE, John SWM. Nicotinamide treatment robustly protects from inherited mouse glaucoma. Commun Integr Biol 2018, 11: e1356956.PubMedPubMedCentralCrossRef Williams PA, Harder JM, Cardozo BH, Foxworth NE, John SWM. Nicotinamide treatment robustly protects from inherited mouse glaucoma. Commun Integr Biol 2018, 11: e1356956.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Tribble JR, Otmani A, Sun S, Ellis SA, Cimaglia G, Vohra R, et al. Nicotinamide provides neuroprotection in glaucoma by protecting against mitochondrial and metabolic dysfunction. Redox Biol 2021, 43: 101988.PubMedPubMedCentralCrossRef Tribble JR, Otmani A, Sun S, Ellis SA, Cimaglia G, Vohra R, et al. Nicotinamide provides neuroprotection in glaucoma by protecting against mitochondrial and metabolic dysfunction. Redox Biol 2021, 43: 101988.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Yu Y, Fedele G, Celardo I, Loh SHY, Martins LM. Parp mutations protect from mitochondrial toxicity in Alzheimer’s disease. Cell Death Dis 2021, 12: 651.PubMedPubMedCentralCrossRef Yu Y, Fedele G, Celardo I, Loh SHY, Martins LM. Parp mutations protect from mitochondrial toxicity in Alzheimer’s disease. Cell Death Dis 2021, 12: 651.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Jia H, Li X, Gao H, Feng Z, Li X, Zhao L, et al. High doses of nicotinamide prevent oxidative mitochondrial dysfunction in a cellular model and improve motor deficit in a Drosophila model of Parkinson’s disease. J Neurosci Res 2008, 86: 2083–2090.PubMedCrossRef Jia H, Li X, Gao H, Feng Z, Li X, Zhao L, et al. High doses of nicotinamide prevent oxidative mitochondrial dysfunction in a cellular model and improve motor deficit in a Drosophila model of Parkinson’s disease. J Neurosci Res 2008, 86: 2083–2090.PubMedCrossRef
68.
Zurück zum Zitat Chen X, Amorim JA, Moustafa GA, Lee JJ, Yu Z, Ishihara K, et al. Neuroprotective effects and mechanisms of action of nicotinamide mononucleotide (NMN) in a photoreceptor degenerative model of retinal detachment. Aging 2020, 12: 24504–24521.PubMedPubMedCentralCrossRef Chen X, Amorim JA, Moustafa GA, Lee JJ, Yu Z, Ishihara K, et al. Neuroprotective effects and mechanisms of action of nicotinamide mononucleotide (NMN) in a photoreceptor degenerative model of retinal detachment. Aging 2020, 12: 24504–24521.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Lee D, Tomita Y, Miwa Y, Shinojima A, Ban N, Yamaguchi S, et al. Nicotinamide mononucleotide prevents retinal dysfunction in a mouse model of retinal ischemia/reperfusion injury. Int J Mol Sci 2022, 23: 11228.PubMedPubMedCentralCrossRef Lee D, Tomita Y, Miwa Y, Shinojima A, Ban N, Yamaguchi S, et al. Nicotinamide mononucleotide prevents retinal dysfunction in a mouse model of retinal ischemia/reperfusion injury. Int J Mol Sci 2022, 23: 11228.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Park JH, Long A, Owens K, Kristian T. Nicotinamide mononucleotide inhibits post-ischemic NAD+ degradation and dramatically ameliorates brain damage following global cerebral ischemia. Neurobiol Dis 2016, 95: 102–110.PubMedPubMedCentralCrossRef Park JH, Long A, Owens K, Kristian T. Nicotinamide mononucleotide inhibits post-ischemic NAD+ degradation and dramatically ameliorates brain damage following global cerebral ischemia. Neurobiol Dis 2016, 95: 102–110.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E, et al. NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A 2018, 115: E1876–E1885.PubMedPubMedCentralCrossRef Hou Y, Lautrup S, Cordonnier S, Wang Y, Croteau DL, Zavala E, et al. NAD+ supplementation normalizes key Alzheimer’s features and DNA damage responses in a new AD mouse model with introduced DNA repair deficiency. Proc Natl Acad Sci U S A 2018, 115: E1876–E1885.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Gong B, Pan Y, Vempati P, Zhao W, Knable L, Ho L, et al. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-γ coactivator 1α regulated β-secretase 1 degradation and mitochondrial gene expression in Alzheimer’s mouse models. Neurobiol Aging 2013, 34: 1581–1588.PubMedPubMedCentralCrossRef Gong B, Pan Y, Vempati P, Zhao W, Knable L, Ho L, et al. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-γ coactivator 1α regulated β-secretase 1 degradation and mitochondrial gene expression in Alzheimer’s mouse models. Neurobiol Aging 2013, 34: 1581–1588.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, et al. NAD+ supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci U S A 2021, 118: e2011226118.PubMedPubMedCentralCrossRef Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, et al. NAD+ supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci U S A 2021, 118: e2011226118.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 2016, 352: 1436–1443.ADSPubMedCrossRef Zhang H, Ryu D, Wu Y, Gariani K, Wang X, Luan P, et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 2016, 352: 1436–1443.ADSPubMedCrossRef
75.
Zurück zum Zitat Vaur P, Brugg B, Mericskay M, Li Z, Schmidt MS, Vivien D, et al. Nicotinamide riboside, a form of vitamin B3, protects against excitotoxicity-induced axonal degeneration. FASEB J 2017, 31: 5440–5452.PubMedCrossRef Vaur P, Brugg B, Mericskay M, Li Z, Schmidt MS, Vivien D, et al. Nicotinamide riboside, a form of vitamin B3, protects against excitotoxicity-induced axonal degeneration. FASEB J 2017, 31: 5440–5452.PubMedCrossRef
76.
Zurück zum Zitat Zhang X, Zhang N, Chrenek MA, Girardot PE, Wang J, Sellers JT, et al. Systemic treatment with nicotinamide riboside is protective in two mouse models of retinal ganglion cell damage. Pharmaceutics 2021, 13: 893.PubMedPubMedCentralCrossRef Zhang X, Zhang N, Chrenek MA, Girardot PE, Wang J, Sellers JT, et al. Systemic treatment with nicotinamide riboside is protective in two mouse models of retinal ganglion cell damage. Pharmaceutics 2021, 13: 893.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Harlan BA, Killoy KM, Pehar M, Liu L, Auwerx J, Vargas MR. Evaluation of the NAD+ biosynthetic pathway in ALS patients and effect of modulating NAD+ levels in hSOD1-linked ALS mouse models. Exp Neurol 2020, 327: 113219.PubMedPubMedCentralCrossRef Harlan BA, Killoy KM, Pehar M, Liu L, Auwerx J, Vargas MR. Evaluation of the NAD+ biosynthetic pathway in ALS patients and effect of modulating NAD+ levels in hSOD1-linked ALS mouse models. Exp Neurol 2020, 327: 113219.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Zhou Q, Zhu L, Qiu W, Liu Y, Yang F, Chen W, et al. Nicotinamide riboside enhances mitochondrial proteostasis and adult neurogenesis through activation of mitochondrial unfolded protein response signaling in the brain of ALS SOD1G93A mice. Int J Biol Sci 2020, 16: 284–297.PubMedPubMedCentralCrossRef Zhou Q, Zhu L, Qiu W, Liu Y, Yang F, Chen W, et al. Nicotinamide riboside enhances mitochondrial proteostasis and adult neurogenesis through activation of mitochondrial unfolded protein response signaling in the brain of ALS SOD1G93A mice. Int J Biol Sci 2020, 16: 284–297.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Cheng YH, Zhao JH, Zong WF, Wei XJ, Xu Z, Yuan Y, et al. Acute treatment with nicotinamide riboside chloride reduces hippocampal damage and preserves the cognitive function of mice with ischemic injury. Neurochem Res 2022, 47: 2244–2253.PubMedCrossRef Cheng YH, Zhao JH, Zong WF, Wei XJ, Xu Z, Yuan Y, et al. Acute treatment with nicotinamide riboside chloride reduces hippocampal damage and preserves the cognitive function of mice with ischemic injury. Neurochem Res 2022, 47: 2244–2253.PubMedCrossRef
80.
Zurück zum Zitat Blaya MO, Bramlett HM, Naidoo J, Pieper AA. Neuroprotective efficacy of a proneurogenic compound after traumatic brain injury. J Neurotrauma 2014, 31: 476–486.PubMedPubMedCentralCrossRef Blaya MO, Bramlett HM, Naidoo J, Pieper AA. Neuroprotective efficacy of a proneurogenic compound after traumatic brain injury. J Neurotrauma 2014, 31: 476–486.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Yin TC, Britt JK, De Jesús-Cortés H, Lu Y, Genova RM, Khan MZ, et al. P7C3 neuroprotective chemicals block axonal degeneration and preserve function after traumatic brain injury. Cell Rep 2014, 8: 1731–1740.PubMedPubMedCentralCrossRef Yin TC, Britt JK, De Jesús-Cortés H, Lu Y, Genova RM, Khan MZ, et al. P7C3 neuroprotective chemicals block axonal degeneration and preserve function after traumatic brain injury. Cell Rep 2014, 8: 1731–1740.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat De Jesús-Cortés H, Miller AD, Britt JK, DeMarco AJ, De Jesús-Cortés M, Stuebing E, et al. Protective efficacy of P7C3-S243 in the 6-hydroxydopamine model of Parkinson’s disease. NPJ Parkinsons Dis 2015, 1: 15010-.PubMedPubMedCentralCrossRef De Jesús-Cortés H, Miller AD, Britt JK, DeMarco AJ, De Jesús-Cortés M, Stuebing E, et al. Protective efficacy of P7C3-S243 in the 6-hydroxydopamine model of Parkinson’s disease. NPJ Parkinsons Dis 2015, 1: 15010-.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Kemp SP, Szynkaruk M, Stanoulis KN, Wood MD, Liu EH, Willand MP, et al. Pharmacologic rescue of motor and sensory function by the neuroprotective compound P7C3 following neonatal nerve injury. Neuroscience 2015, 284: 202–216.PubMedCrossRef Kemp SP, Szynkaruk M, Stanoulis KN, Wood MD, Liu EH, Willand MP, et al. Pharmacologic rescue of motor and sensory function by the neuroprotective compound P7C3 following neonatal nerve injury. Neuroscience 2015, 284: 202–216.PubMedCrossRef
84.
Zurück zum Zitat LoCoco PM, Risinger AL, Smith HR, Chavera TS, Berg KA, Clarke WP. Pharmacological augmentation of nicotinamide phosphoribosyltransferase (NAMPT) protects against paclitaxel-induced peripheral neuropathy. Elife 2017, 6: e29626.PubMedPubMedCentralCrossRef LoCoco PM, Risinger AL, Smith HR, Chavera TS, Berg KA, Clarke WP. Pharmacological augmentation of nicotinamide phosphoribosyltransferase (NAMPT) protects against paclitaxel-induced peripheral neuropathy. Elife 2017, 6: e29626.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Loris ZB, Hynton JR, Pieper AA, Dietrich WD. Beneficial effects of delayed P7C3-A20 treatment after transient MCAO in rats. Transl Stroke Res 2018, 9: 146–156.PubMedCrossRef Loris ZB, Hynton JR, Pieper AA, Dietrich WD. Beneficial effects of delayed P7C3-A20 treatment after transient MCAO in rats. Transl Stroke Res 2018, 9: 146–156.PubMedCrossRef
86.
Zurück zum Zitat Yao H, Liu M, Wang L, Zu Y, Wu C, Li C, et al. Discovery of small-molecule activators of nicotinamide phosphoribosyltransferase (NAMPT) and their preclinical neuroprotective activity. Cell Res 2022, 32: 570–584.PubMedPubMedCentralCrossRef Yao H, Liu M, Wang L, Zu Y, Wu C, Li C, et al. Discovery of small-molecule activators of nicotinamide phosphoribosyltransferase (NAMPT) and their preclinical neuroprotective activity. Cell Res 2022, 32: 570–584.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Kim HJ, Cao W, Oh GS, Lee S, Shen A, Khadka D, et al. Augmentation of cellular NAD+ by NQO1 enzymatic action improves age-related hearing impairment. Aging Cell 2019, 18: e13016.PubMedPubMedCentralCrossRef Kim HJ, Cao W, Oh GS, Lee S, Shen A, Khadka D, et al. Augmentation of cellular NAD+ by NQO1 enzymatic action improves age-related hearing impairment. Aging Cell 2019, 18: e13016.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Kim HJ, Oh GS, Shen A, Lee SB, Choe SK, Kwon KB, et al. Augmentation of NAD(+) by NQO1 attenuates cisplatin-mediated hearing impairment. Cell Death Dis 2014, 5: e1292.PubMedPubMedCentralCrossRef Kim HJ, Oh GS, Shen A, Lee SB, Choe SK, Kwon KB, et al. Augmentation of NAD(+) by NQO1 attenuates cisplatin-mediated hearing impairment. Cell Death Dis 2014, 5: e1292.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Escande C, Nin V, Price NL, Capellini V, Gomes AP, Barbosa MT, et al. Flavonoid apigenin is an inhibitor of the NAD+ ase CD38: Implications for cellular NAD+ metabolism, protein acetylation, and treatment of metabolic syndrome. Diabetes 2013, 62: 1084–1093.PubMedPubMedCentralCrossRef Escande C, Nin V, Price NL, Capellini V, Gomes AP, Barbosa MT, et al. Flavonoid apigenin is an inhibitor of the NAD+ ase CD38: Implications for cellular NAD+ metabolism, protein acetylation, and treatment of metabolic syndrome. Diabetes 2013, 62: 1084–1093.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Ahmedy OA, Abdelghany TM, El-Shamarka MEA, Khattab MA, El-Tanbouly DM. Apigenin attenuates LPS-induced neurotoxicity and cognitive impairment in mice via promoting mitochondrial fusion/mitophagy: Role of SIRT3/PINK1/Parkin pathway. Psychopharmacology 2022, 239: 3903–3917.PubMedPubMedCentralCrossRef Ahmedy OA, Abdelghany TM, El-Shamarka MEA, Khattab MA, El-Tanbouly DM. Apigenin attenuates LPS-induced neurotoxicity and cognitive impairment in mice via promoting mitochondrial fusion/mitophagy: Role of SIRT3/PINK1/Parkin pathway. Psychopharmacology 2022, 239: 3903–3917.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Watanabe R, Kurose T, Morishige Y, Fujimori K. Protective effects of fisetin against 6-OHDA-induced apoptosis by activation of PI3K-Akt signaling in human neuroblastoma SH-SY5Y cells. Neurochem Res 2018, 43: 488–499.PubMedCrossRef Watanabe R, Kurose T, Morishige Y, Fujimori K. Protective effects of fisetin against 6-OHDA-induced apoptosis by activation of PI3K-Akt signaling in human neuroblastoma SH-SY5Y cells. Neurochem Res 2018, 43: 488–499.PubMedCrossRef
92.
Zurück zum Zitat Geraets L, Moonen HJJ, Brauers K, Wouters EFM, Bast A, Hageman GJ. Dietary flavones and flavonoles are inhibitors of poly(ADP-ribose)polymerase-1 in pulmonary epithelial cells. J Nutr 2007, 137: 2190–2195.PubMedCrossRef Geraets L, Moonen HJJ, Brauers K, Wouters EFM, Bast A, Hageman GJ. Dietary flavones and flavonoles are inhibitors of poly(ADP-ribose)polymerase-1 in pulmonary epithelial cells. J Nutr 2007, 137: 2190–2195.PubMedCrossRef
93.
Zurück zum Zitat Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 2003, 425: 191–196.ADSPubMedCrossRef Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 2003, 425: 191–196.ADSPubMedCrossRef
94.
Zurück zum Zitat Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol 2015, 40: 134–141.PubMedCrossRef Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol 2015, 40: 134–141.PubMedCrossRef
95.
Zurück zum Zitat Kanai M, Funakoshi H, Takahashi H, Hayakawa T, Mizuno S, Matsumoto K, et al. Tryptophan 2, 3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice. Mol Brain 2009, 2: 8.PubMedPubMedCentralCrossRef Kanai M, Funakoshi H, Takahashi H, Hayakawa T, Mizuno S, Matsumoto K, et al. Tryptophan 2, 3-dioxygenase is a key modulator of physiological neurogenesis and anxiety-related behavior in mice. Mol Brain 2009, 2: 8.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Yu D, Tao BB, Yang YY, Du LS, Yang SS, He XJ, et al. The IDO inhibitor coptisine ameliorates cognitive impairment in a mouse model of Alzheimer’s disease. J Alzheimers Dis 2015, 43: 291–302.PubMedCrossRef Yu D, Tao BB, Yang YY, Du LS, Yang SS, He XJ, et al. The IDO inhibitor coptisine ameliorates cognitive impairment in a mouse model of Alzheimer’s disease. J Alzheimers Dis 2015, 43: 291–302.PubMedCrossRef
97.
Zurück zum Zitat Woodling NS, Colas D, Wang Q, Minhas P, Panchal M, Liang X, et al. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer’s disease model mice. Brain 2016, 139: 2063–2081.PubMedPubMedCentralCrossRef Woodling NS, Colas D, Wang Q, Minhas P, Panchal M, Liang X, et al. Cyclooxygenase inhibition targets neurons to prevent early behavioural decline in Alzheimer’s disease model mice. Brain 2016, 139: 2063–2081.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Oxenkrug GF, Navrotskaya V, Voroboyva L, Summergrad P. Extension of life span of Drosophila melanogaster by the inhibitors of tryptophan-kynurenine metabolism. Fly 2011, 5: 307–309.PubMedPubMedCentralCrossRef Oxenkrug GF, Navrotskaya V, Voroboyva L, Summergrad P. Extension of life span of Drosophila melanogaster by the inhibitors of tryptophan-kynurenine metabolism. Fly 2011, 5: 307–309.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Opitz CA, Heiland I. Dynamics of NAD-metabolism: Everything but constant. Biochem Soc Trans 2015, 43: 1127–1132.PubMedCrossRef Opitz CA, Heiland I. Dynamics of NAD-metabolism: Everything but constant. Biochem Soc Trans 2015, 43: 1127–1132.PubMedCrossRef
100.
Zurück zum Zitat Platten M, Nollen EAA, Röhrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov 2019, 18: 379–401.PubMedCrossRef Platten M, Nollen EAA, Röhrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov 2019, 18: 379–401.PubMedCrossRef
101.
Zurück zum Zitat Liu L, Su X, Quinn WJ 3rd, Hui S, Krukenberg K, Frederick DW, et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab 2018, 27: 1067-1080.e5.PubMedPubMedCentralCrossRef Liu L, Su X, Quinn WJ 3rd, Hui S, Krukenberg K, Frederick DW, et al. Quantitative analysis of NAD synthesis-breakdown fluxes. Cell Metab 2018, 27: 1067-1080.e5.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Gasperi V, Sibilano M, Savini I, Catani MV. Niacin in the central nervous system: An update of biological aspects and clinical applications. Int J Mol Sci 2019, 20: 974.PubMedPubMedCentralCrossRef Gasperi V, Sibilano M, Savini I, Catani MV. Niacin in the central nervous system: An update of biological aspects and clinical applications. Int J Mol Sci 2019, 20: 974.PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Pirinen E, Auranen M, Khan NA, Brilhante V, Urho N, Pessia A, et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab 2020, 32: 144.PubMedCrossRef Pirinen E, Auranen M, Khan NA, Brilhante V, Urho N, Pessia A, et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab 2020, 32: 144.PubMedCrossRef
104.
Zurück zum Zitat Investigators AH, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011, 365: 2255–2267.CrossRef Investigators AH, Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, et al. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011, 365: 2255–2267.CrossRef
105.
Zurück zum Zitat HPS2-THRIVE Collaborative Group. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: Trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J 2013, 34: 1279–1291.PubMedCentralCrossRef HPS2-THRIVE Collaborative Group. HPS2-THRIVE randomized placebo-controlled trial in 25 673 high-risk patients of ER niacin/laropiprant: Trial design, pre-specified muscle and liver outcomes, and reasons for stopping study treatment. Eur Heart J 2013, 34: 1279–1291.PubMedCentralCrossRef
106.
Zurück zum Zitat Ramanathan C, Lackie T, Williams DH, Simone PS, Zhang Y, Bloomer RJ. Oral administration of nicotinamide mononucleotide increases nicotinamide adenine dinucleotide level in an animal brain. Nutrients 2022, 14: 300.PubMedPubMedCentralCrossRef Ramanathan C, Lackie T, Williams DH, Simone PS, Zhang Y, Bloomer RJ. Oral administration of nicotinamide mononucleotide increases nicotinamide adenine dinucleotide level in an animal brain. Nutrients 2022, 14: 300.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, Schultz MB, et al. Impairment of an Endothelial NAD+-H2S Signaling Network Is a Reversible Cause of Vascular Aging. Cell. 2018, 173: 74–89. e20.PubMedPubMedCentralCrossRef Das A, Huang GX, Bonkowski MS, Longchamp A, Li C, Schultz MB, et al. Impairment of an Endothelial NAD+-H2S Signaling Network Is a Reversible Cause of Vascular Aging. Cell. 2018, 173: 74–89. e20.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Kiss T, Nyúl-Tóth Á, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, et al. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: Transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Gero Science 2020, 42: 527–546.PubMedPubMedCentral Kiss T, Nyúl-Tóth Á, Balasubramanian P, Tarantini S, Ahire C, Yabluchanskiy A, et al. Nicotinamide mononucleotide (NMN) supplementation promotes neurovascular rejuvenation in aged mice: Transcriptional footprint of SIRT1 activation, mitochondrial protection, anti-inflammatory, and anti-apoptotic effects. Gero Science 2020, 42: 527–546.PubMedPubMedCentral
109.
Zurück zum Zitat Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab 2016, 24: 795–806.PubMedPubMedCentralCrossRef Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, et al. Long-term administration of nicotinamide mononucleotide mitigates age-associated physiological decline in mice. Cell Metab 2016, 24: 795–806.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell 2016, 15: 522–530.PubMedPubMedCentralCrossRef de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills KF, et al. Nicotinamide mononucleotide supplementation reverses vascular dysfunction and oxidative stress with aging in mice. Aging Cell 2016, 15: 522–530.PubMedPubMedCentralCrossRef
111.
112.
Zurück zum Zitat Zheng SL, Wang DS, Dong X, Guan YF, Qi Q, Hu WJ, et al. Distribution of nicotinamide mononucleotide after intravenous injection in normal and ischemic stroke mice. Curr Pharm Biotechnol 2023, 24: 299–309.PubMedCrossRef Zheng SL, Wang DS, Dong X, Guan YF, Qi Q, Hu WJ, et al. Distribution of nicotinamide mononucleotide after intravenous injection in normal and ischemic stroke mice. Curr Pharm Biotechnol 2023, 24: 299–309.PubMedCrossRef
113.
Zurück zum Zitat Pencina KM, Lavu S, Dos Santos M, Beleva YM, Cheng M, Livingston D, et al. MIB-626, an oral formulation of a microcrystalline unique polymorph of β-nicotinamide mononucleotide, increases circulating nicotinamide adenine dinucleotide and its metabolome in middle-aged and older adults. J Gerontol A Biol Sci Med Sci 2023, 78: 90–96.PubMedCrossRef Pencina KM, Lavu S, Dos Santos M, Beleva YM, Cheng M, Livingston D, et al. MIB-626, an oral formulation of a microcrystalline unique polymorph of β-nicotinamide mononucleotide, increases circulating nicotinamide adenine dinucleotide and its metabolome in middle-aged and older adults. J Gerontol A Biol Sci Med Sci 2023, 78: 90–96.PubMedCrossRef
114.
Zurück zum Zitat Niu KM, Bao T, Gao L, Ru M, Li Y, Jiang L, et al. The impacts of short-term NMN supplementation on serum metabolism, fecal microbiota, and telomere length in pre-aging phase. Front Nutr 2021, 8: 756243.PubMedPubMedCentralCrossRef Niu KM, Bao T, Gao L, Ru M, Li Y, Jiang L, et al. The impacts of short-term NMN supplementation on serum metabolism, fecal microbiota, and telomere length in pre-aging phase. Front Nutr 2021, 8: 756243.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Schmidt MS, Brenner C. Absence of evidence that Slc12a8 encodes a nicotinamide mononucleotide transporter. Nat Metab 2019, 1: 660–661.PubMedCrossRef Schmidt MS, Brenner C. Absence of evidence that Slc12a8 encodes a nicotinamide mononucleotide transporter. Nat Metab 2019, 1: 660–661.PubMedCrossRef
116.
Zurück zum Zitat Grozio A, Mills KF, Yoshino J, Bruzzone S, Sociali G, Tokizane K, et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat Metab 2019, 1: 47–57.PubMedPubMedCentralCrossRef Grozio A, Mills KF, Yoshino J, Bruzzone S, Sociali G, Tokizane K, et al. Slc12a8 is a nicotinamide mononucleotide transporter. Nat Metab 2019, 1: 47–57.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Ratajczak J, Joffraud M, Trammell SAJ, Ras R, Canela N, Boutant M, et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nat Commun 2016, 7: 13103.ADSPubMedPubMedCentralCrossRef Ratajczak J, Joffraud M, Trammell SAJ, Ras R, Canela N, Boutant M, et al. NRK1 controls nicotinamide mononucleotide and nicotinamide riboside metabolism in mammalian cells. Nat Commun 2016, 7: 13103.ADSPubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Brakedal B, Dölle C, Riemer F, Ma Y, Nido GS, Skeie GO, et al. The NADPARK study: A randomized phase I trial of nicotinamide riboside supplementation in Parkinson’s disease. Cell Metab 2022, 34: 396-407.e6.PubMedCrossRef Brakedal B, Dölle C, Riemer F, Ma Y, Nido GS, Skeie GO, et al. The NADPARK study: A randomized phase I trial of nicotinamide riboside supplementation in Parkinson’s disease. Cell Metab 2022, 34: 396-407.e6.PubMedCrossRef
119.
Zurück zum Zitat Schöndorf DC, Ivanyuk D, Baden P, Sanchez-Martinez A, De Cicco S, Yu C, et al. The NAD+ precursor nicotinamide riboside rescues mitochondrial defects and neuronal loss in iPSC and fly models of parkinson’s disease. Cell Rep 2018, 23: 2976–2988.PubMedCrossRef Schöndorf DC, Ivanyuk D, Baden P, Sanchez-Martinez A, De Cicco S, Yu C, et al. The NAD+ precursor nicotinamide riboside rescues mitochondrial defects and neuronal loss in iPSC and fly models of parkinson’s disease. Cell Rep 2018, 23: 2976–2988.PubMedCrossRef
120.
Zurück zum Zitat Giroud-Gerbetant J, Joffraud M, Giner MP, Cercillieux A, Bartova S, Makarov MV, et al. A reduced form of nicotinamide riboside defines a new path for NAD+ biosynthesis and acts as an orally bioavailable NAD+ precursor. Mol Metab 2019, 30: 192–202.PubMedPubMedCentralCrossRef Giroud-Gerbetant J, Joffraud M, Giner MP, Cercillieux A, Bartova S, Makarov MV, et al. A reduced form of nicotinamide riboside defines a new path for NAD+ biosynthesis and acts as an orally bioavailable NAD+ precursor. Mol Metab 2019, 30: 192–202.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Khaidizar FD, Bessho Y, Nakahata Y. Nicotinamide phosphoribosyltransferase as a key molecule of the aging/senescence process. Int J Mol Sci 2021, 22: 3709.PubMedPubMedCentralCrossRef Khaidizar FD, Bessho Y, Nakahata Y. Nicotinamide phosphoribosyltransferase as a key molecule of the aging/senescence process. Int J Mol Sci 2021, 22: 3709.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Hosseini L, Mahmoudi J, Pashazadeh F, Salehi-Pourmehr H, Sadigh-Eteghad S. Protective effects of nicotinamide adenine dinucleotide and related precursors in alzheimer’s disease: A systematic review of preclinical studies. J Mol Neurosci 2021, 71: 1425–1435.PubMedCrossRef Hosseini L, Mahmoudi J, Pashazadeh F, Salehi-Pourmehr H, Sadigh-Eteghad S. Protective effects of nicotinamide adenine dinucleotide and related precursors in alzheimer’s disease: A systematic review of preclinical studies. J Mol Neurosci 2021, 71: 1425–1435.PubMedCrossRef
123.
Zurück zum Zitat Wang G, Han T, Nijhawan D, Theodoropoulos P, Naidoo J, Yadavalli S, et al. P7C3 neuroprotective chemicals function by activating the rate-limiting enzyme in NAD salvage. Cell 2014, 158: 1324–1334.PubMedPubMedCentralCrossRef Wang G, Han T, Nijhawan D, Theodoropoulos P, Naidoo J, Yadavalli S, et al. P7C3 neuroprotective chemicals function by activating the rate-limiting enzyme in NAD salvage. Cell 2014, 158: 1324–1334.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Pieper AA, McKnight SL, Ready JM. P7C3 and an unbiased approach to drug discovery for neurodegenerative diseases. Chem Soc Rev 2014, 43: 6716–6726.PubMedPubMedCentralCrossRef Pieper AA, McKnight SL, Ready JM. P7C3 and an unbiased approach to drug discovery for neurodegenerative diseases. Chem Soc Rev 2014, 43: 6716–6726.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Latchney SE, Jaramillo TC, Rivera PD, Eisch AJ, Powell CM. Chronic P7C3 treatment restores hippocampal neurogenesis in the Ts65Dn mouse model of Down Syndrome[Corrected. Neurosci Lett 2015, 591: 86–92.PubMedPubMedCentralCrossRef Latchney SE, Jaramillo TC, Rivera PD, Eisch AJ, Powell CM. Chronic P7C3 treatment restores hippocampal neurogenesis in the Ts65Dn mouse model of Down Syndrome[Corrected. Neurosci Lett 2015, 591: 86–92.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Bauman MD, Schumann CM, Carlson EL, Taylor SL, Vázquez-Rosa E, Cintrón-Pérez CJ, et al. Neuroprotective efficacy of P7C3 compounds in primate hippocampus. Transl Psychiatry 2018, 8: 202.PubMedPubMedCentralCrossRef Bauman MD, Schumann CM, Carlson EL, Taylor SL, Vázquez-Rosa E, Cintrón-Pérez CJ, et al. Neuroprotective efficacy of P7C3 compounds in primate hippocampus. Transl Psychiatry 2018, 8: 202.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Vázquez-Rosa E, Shin MK, Dhar M, Chaubey K, Cintrón-Pérez CJ, Tang X, et al. P7C3-A20 treatment one year after TBI in mice repairs the blood-brain barrier, arrests chronic neurodegeneration, and restores cognition. Proc Natl Acad Sci U S A 2020, 117: 27667–27675.ADSPubMedPubMedCentralCrossRef Vázquez-Rosa E, Shin MK, Dhar M, Chaubey K, Cintrón-Pérez CJ, Tang X, et al. P7C3-A20 treatment one year after TBI in mice repairs the blood-brain barrier, arrests chronic neurodegeneration, and restores cognition. Proc Natl Acad Sci U S A 2020, 117: 27667–27675.ADSPubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Tur J, Badole SL, Manickam R, Chapalamadugu KC, Xuan W, Guida W, et al. Cardioprotective effects of 1-(3, 6-dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol in diabetic hearts via nicotinamide phosphoribosyltransferase activation. J Pharmacol Exp Ther 2022, 382: 233–245.PubMedPubMedCentralCrossRef Tur J, Badole SL, Manickam R, Chapalamadugu KC, Xuan W, Guida W, et al. Cardioprotective effects of 1-(3, 6-dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol in diabetic hearts via nicotinamide phosphoribosyltransferase activation. J Pharmacol Exp Ther 2022, 382: 233–245.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Manickam R, Tur J, Badole SL, Chapalamadugu KC, Sinha P, Wang Z, et al. Nampt activator P7C3 ameliorates diabetes and improves skeletal muscle function modulating cell metabolism and lipid mediators. J Cachexia Sarcopenia Muscle 2022, 13: 1177–1196.PubMedPubMedCentralCrossRef Manickam R, Tur J, Badole SL, Chapalamadugu KC, Sinha P, Wang Z, et al. Nampt activator P7C3 ameliorates diabetes and improves skeletal muscle function modulating cell metabolism and lipid mediators. J Cachexia Sarcopenia Muscle 2022, 13: 1177–1196.PubMedPubMedCentralCrossRef
130.
Zurück zum Zitat Gardell SJ, Hopf M, Khan A, Dispagna M, Hampton Sessions E, Falter R, et al. Boosting NAD+ with a small molecule that activates NAMPT. Nat Commun 2019, 10: 3241.ADSPubMedPubMedCentralCrossRef Gardell SJ, Hopf M, Khan A, Dispagna M, Hampton Sessions E, Falter R, et al. Boosting NAD+ with a small molecule that activates NAMPT. Nat Commun 2019, 10: 3241.ADSPubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Pinkerton AB, Hershberger P, Maloney PR, Peddibhotla S, Hopf M, et al. Optimization of a urea-containing series of nicotinamide phosphoribosyltransferase (NAMPT) activators. Bioorg Med Chem Lett 2021, 41: 128007.PubMedCrossRef Pinkerton AB, Hershberger P, Maloney PR, Peddibhotla S, Hopf M, et al. Optimization of a urea-containing series of nicotinamide phosphoribosyltransferase (NAMPT) activators. Bioorg Med Chem Lett 2021, 41: 128007.PubMedCrossRef
132.
Zurück zum Zitat Akiu M, Tsuji T, Sogawa Y, Terayama K, Yokoyama M, Tanaka J, et al. Discovery of 1-[2-(1-methyl-1H-pyrazol-5-yl)-[1, 2, 4]triazolo[1, 5-a]pyridin-6-yl]-3-(pyridin-4-ylmethyl)urea as a potent NAMPT (nicotinamide phosphoribosyltransferase) activator with attenuated CYP inhibition. Bioorg Med Chem Lett 2021, 43: 128048.PubMedCrossRef Akiu M, Tsuji T, Sogawa Y, Terayama K, Yokoyama M, Tanaka J, et al. Discovery of 1-[2-(1-methyl-1H-pyrazol-5-yl)-[1, 2, 4]triazolo[1, 5-a]pyridin-6-yl]-3-(pyridin-4-ylmethyl)urea as a potent NAMPT (nicotinamide phosphoribosyltransferase) activator with attenuated CYP inhibition. Bioorg Med Chem Lett 2021, 43: 128048.PubMedCrossRef
133.
Zurück zum Zitat Wang L, Liu M, Zu Y, Yao H, Wu C, Zhang R, et al. Optimization of NAMPT activators to achieve in vivo neuroprotective efficacy. Eur J Med Chem 2022, 236: 114260.PubMedCrossRef Wang L, Liu M, Zu Y, Yao H, Wu C, Zhang R, et al. Optimization of NAMPT activators to achieve in vivo neuroprotective efficacy. Eur J Med Chem 2022, 236: 114260.PubMedCrossRef
134.
Zurück zum Zitat Sociali G, Grozio A, Caffa I, Schuster S, Becherini P, Damonte P, et al. SIRT6 deacetylase activity regulates NAMPT activity and NAD(P)(H) pools in cancer cells. FASEB J 2019, 33: 3704–3717.PubMedCrossRef Sociali G, Grozio A, Caffa I, Schuster S, Becherini P, Damonte P, et al. SIRT6 deacetylase activity regulates NAMPT activity and NAD(P)(H) pools in cancer cells. FASEB J 2019, 33: 3704–3717.PubMedCrossRef
135.
Zurück zum Zitat Collier JB, Schnellmann RG. Extracellular signal-regulated kinase 1/2 regulates NAD metabolism during acute kidney injury through microRNA-34a-mediated NAMPT expression. Cell Mol Life Sci 2020, 77: 3643–3655.PubMedCrossRef Collier JB, Schnellmann RG. Extracellular signal-regulated kinase 1/2 regulates NAD metabolism during acute kidney injury through microRNA-34a-mediated NAMPT expression. Cell Mol Life Sci 2020, 77: 3643–3655.PubMedCrossRef
136.
Zurück zum Zitat Xie W, Zhu T, Zhou P, Xu H, Meng X, Ding T, et al. Notoginseng leaf triterpenes ameliorates OGD/R-induced neuronal injury via SIRT1/2/3-Foxo3a-MnSOD/PGC-1 α signaling pathways mediated by the NAMPT-NAD pathway. Oxid Med Cell Longev 2020, 2020: 7308386.PubMedPubMedCentralCrossRef Xie W, Zhu T, Zhou P, Xu H, Meng X, Ding T, et al. Notoginseng leaf triterpenes ameliorates OGD/R-induced neuronal injury via SIRT1/2/3-Foxo3a-MnSOD/PGC-1 α signaling pathways mediated by the NAMPT-NAD pathway. Oxid Med Cell Longev 2020, 2020: 7308386.PubMedPubMedCentralCrossRef
137.
138.
Zurück zum Zitat Ali YO, Li-Kroeger D, Bellen HJ, Zhai RG, Lu HC. NMNATs, evolutionarily conserved neuronal maintenance factors. Trends Neurosci 2013, 36: 632–640.PubMedCrossRef Ali YO, Li-Kroeger D, Bellen HJ, Zhai RG, Lu HC. NMNATs, evolutionarily conserved neuronal maintenance factors. Trends Neurosci 2013, 36: 632–640.PubMedCrossRef
139.
Zurück zum Zitat Wu X, Hu F, Zeng J, Han L, Qiu D, Wang H, et al. NMNAT2-mediated NAD+ generation is essential for quality control of aged oocytes. Aging Cell 2019, 18: e12955.PubMedPubMedCentralCrossRef Wu X, Hu F, Zeng J, Han L, Qiu D, Wang H, et al. NMNAT2-mediated NAD+ generation is essential for quality control of aged oocytes. Aging Cell 2019, 18: e12955.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Ali YO, Allen HM, Yu L, Li-Kroeger D, Bakhshizadehmahmoudi D, Hatcher A, et al. NMNAT2: HSP90 complex mediates proteostasis in proteinopathies. PLoS Biol 2016, 14: e1002472.PubMedPubMedCentralCrossRef Ali YO, Allen HM, Yu L, Li-Kroeger D, Bakhshizadehmahmoudi D, Hatcher A, et al. NMNAT2: HSP90 complex mediates proteostasis in proteinopathies. PLoS Biol 2016, 14: e1002472.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Cheng XS, Zhao KP, Jiang X, Du LL, Li XH, Ma ZW, et al. Nmnat2 attenuates tau phosphorylation through activation of PP2A. J Alzheimers Dis 2013, 36: 185–195.PubMedCrossRef Cheng XS, Zhao KP, Jiang X, Du LL, Li XH, Ma ZW, et al. Nmnat2 attenuates tau phosphorylation through activation of PP2A. J Alzheimers Dis 2013, 36: 185–195.PubMedCrossRef
142.
Zurück zum Zitat Cai Y, Yu SS, Chen SR, Pi RB, Gao S, Li H, et al. Nmnat2 protects cardiomyocytes from hypertrophy via activation of SIRT6. FEBS Lett 2012, 586: 866–874.PubMedCrossRef Cai Y, Yu SS, Chen SR, Pi RB, Gao S, Li H, et al. Nmnat2 protects cardiomyocytes from hypertrophy via activation of SIRT6. FEBS Lett 2012, 586: 866–874.PubMedCrossRef
143.
Zurück zum Zitat Rossi F, Geiszler PC, Meng W, Barron MR, Prior M, Herd-Smith A, et al. NAD-biosynthetic enzyme NMNAT1 reduces early behavioral impairment in the htau mouse model of tauopathy. Behav Brain Res 2018, 339: 140–152.PubMedPubMedCentralCrossRef Rossi F, Geiszler PC, Meng W, Barron MR, Prior M, Herd-Smith A, et al. NAD-biosynthetic enzyme NMNAT1 reduces early behavioral impairment in the htau mouse model of tauopathy. Behav Brain Res 2018, 339: 140–152.PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Fang F, Zhuang P, Feng X, Liu P, Liu D, Huang H, et al. NMNAT2 is downregulated in glaucomatous RGCs, and RGC-specific gene therapy rescues neurodegeneration and visual function. Mol Ther 2022, 30: 1421–1431.PubMedPubMedCentralCrossRef Fang F, Zhuang P, Feng X, Liu P, Liu D, Huang H, et al. NMNAT2 is downregulated in glaucomatous RGCs, and RGC-specific gene therapy rescues neurodegeneration and visual function. Mol Ther 2022, 30: 1421–1431.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Haubrich BA, Ramesha C, Swinney DC. Development of a bioluminescent high-throughput screening assay for nicotinamide mononucleotide adenylyltransferase (NMNAT). SLAS Discov 2020, 25: 33–42.PubMedCrossRef Haubrich BA, Ramesha C, Swinney DC. Development of a bioluminescent high-throughput screening assay for nicotinamide mononucleotide adenylyltransferase (NMNAT). SLAS Discov 2020, 25: 33–42.PubMedCrossRef
146.
Zurück zum Zitat Diaz-Ruiz A, Lanasa M, Garcia J, Mora H, Fan F, Martin-Montalvo A, et al. Overexpression of CYB5R3 and NQO1, two NAD+-producing enzymes, mimics aspects of caloric restriction. Aging Cell 2018, 17: e12767.PubMedPubMedCentralCrossRef Diaz-Ruiz A, Lanasa M, Garcia J, Mora H, Fan F, Martin-Montalvo A, et al. Overexpression of CYB5R3 and NQO1, two NAD+-producing enzymes, mimics aspects of caloric restriction. Aging Cell 2018, 17: e12767.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Pandit A, Kim HJ, Oh GS, Shen A, Lee SB, Khadka D, et al. Dunnione ameliorates cisplatin-induced small intestinal damage by modulating NAD(+) metabolism. Biochem Biophys Res Commun 2015, 467: 697–703.PubMedCrossRef Pandit A, Kim HJ, Oh GS, Shen A, Lee SB, Khadka D, et al. Dunnione ameliorates cisplatin-induced small intestinal damage by modulating NAD(+) metabolism. Biochem Biophys Res Commun 2015, 467: 697–703.PubMedCrossRef
148.
Zurück zum Zitat Khadka D, Kim HJ, Oh GS, Shen A, Lee S, Lee SB, et al. Augmentation of NAD+ levels by enzymatic action of NAD(P)H quinone oxidoreductase 1 attenuates adriamycin-induced cardiac dysfunction in mice. J Mol Cell Cardiol 2018, 124: 45–57.PubMedCrossRef Khadka D, Kim HJ, Oh GS, Shen A, Lee S, Lee SB, et al. Augmentation of NAD+ levels by enzymatic action of NAD(P)H quinone oxidoreductase 1 attenuates adriamycin-induced cardiac dysfunction in mice. J Mol Cell Cardiol 2018, 124: 45–57.PubMedCrossRef
149.
Zurück zum Zitat Nazari Soltan Ahmad S, Rashtchizadeh N, Argani H, Roshangar L, Ghorbani Haghjo A, Sanajou D, et al. Dunnione protects against experimental cisplatin-induced nephrotoxicity by modulating NQO1 and NAD+ levels. Free Radic Res 2018, 52: 808–817.PubMedCrossRef Nazari Soltan Ahmad S, Rashtchizadeh N, Argani H, Roshangar L, Ghorbani Haghjo A, Sanajou D, et al. Dunnione protects against experimental cisplatin-induced nephrotoxicity by modulating NQO1 and NAD+ levels. Free Radic Res 2018, 52: 808–817.PubMedCrossRef
150.
Zurück zum Zitat Oh GS, Lee SB, Karna A, Kim HJ, Shen A, Pandit A, et al. Increased cellular NAD+ level through NQO1 enzymatic action has protective effects on bleomycin-induced lung fibrosis in mice. Tuberc Respir Dis (Seoul) 2016, 79: 257–266.PubMedCrossRef Oh GS, Lee SB, Karna A, Kim HJ, Shen A, Pandit A, et al. Increased cellular NAD+ level through NQO1 enzymatic action has protective effects on bleomycin-induced lung fibrosis in mice. Tuberc Respir Dis (Seoul) 2016, 79: 257–266.PubMedCrossRef
151.
Zurück zum Zitat Talla V, Koilkonda R, Guy J. Gene therapy with single-subunit yeast NADH-ubiquinone oxidoreductase (NDI1) improves the visual function in experimental autoimmune encephalomyelitis (EAE) mice model of multiple sclerosis (MS). Mol Neurobiol 2020, 57: 1952–1965.PubMedCrossRef Talla V, Koilkonda R, Guy J. Gene therapy with single-subunit yeast NADH-ubiquinone oxidoreductase (NDI1) improves the visual function in experimental autoimmune encephalomyelitis (EAE) mice model of multiple sclerosis (MS). Mol Neurobiol 2020, 57: 1952–1965.PubMedCrossRef
152.
Zurück zum Zitat Talla V, Yu H, Chou TH, Porciatti V, Chiodo V, Boye SL, et al. NADH-dehydrogenase type-2 suppresses irreversible visual loss and neurodegeneration in the EAE animal model of MS. Mol Ther 2013, 21: 1876–1888.PubMedPubMedCentralCrossRef Talla V, Yu H, Chou TH, Porciatti V, Chiodo V, Boye SL, et al. NADH-dehydrogenase type-2 suppresses irreversible visual loss and neurodegeneration in the EAE animal model of MS. Mol Ther 2013, 21: 1876–1888.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Han G, Zhen W, Dai Y, Yu H, Li D, Ma T. Dihuang-yinzi alleviates cognition deficits via targeting energy-related metabolism in an alzheimer mouse model as demonstrated by integration of metabolomics and network pharmacology. Front Aging Neurosci 2022, 14: 873929.PubMedPubMedCentralCrossRef Han G, Zhen W, Dai Y, Yu H, Li D, Ma T. Dihuang-yinzi alleviates cognition deficits via targeting energy-related metabolism in an alzheimer mouse model as demonstrated by integration of metabolomics and network pharmacology. Front Aging Neurosci 2022, 14: 873929.PubMedPubMedCentralCrossRef
154.
155.
Zurück zum Zitat Apiraksattayakul S, Pingaew R, Prachayasittikul V, Ruankham W, Jongwachirachai P, Songtawee N, et al. Neuroprotective properties of bis-sulfonamide derivatives against 6-OHDA-induced parkinson’s model via sirtuin 1 activity and in silico pharmacokinetic properties. Front Mol Neurosci 2022, 15: 890838.PubMedPubMedCentralCrossRef Apiraksattayakul S, Pingaew R, Prachayasittikul V, Ruankham W, Jongwachirachai P, Songtawee N, et al. Neuroprotective properties of bis-sulfonamide derivatives against 6-OHDA-induced parkinson’s model via sirtuin 1 activity and in silico pharmacokinetic properties. Front Mol Neurosci 2022, 15: 890838.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Romeo-Guitart D, Marcos-DeJuana C, Marmolejo-Martínez-Artesero S, Navarro X, Casas C. Novel neuroprotective therapy with NeuroHeal by autophagy induction for damaged neonatal motoneurons. Theranostics 2020, 10: 5154–5168.PubMedPubMedCentralCrossRef Romeo-Guitart D, Marcos-DeJuana C, Marmolejo-Martínez-Artesero S, Navarro X, Casas C. Novel neuroprotective therapy with NeuroHeal by autophagy induction for damaged neonatal motoneurons. Theranostics 2020, 10: 5154–5168.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-dependent deacetylase SIRT2 protects mouse brain against ischemic stroke. Mol Neurobiol 2017, 54: 7251–7261.PubMedCrossRef Xie XQ, Zhang P, Tian B, Chen XQ. Downregulation of NAD-dependent deacetylase SIRT2 protects mouse brain against ischemic stroke. Mol Neurobiol 2017, 54: 7251–7261.PubMedCrossRef
158.
Zurück zum Zitat Mellini P, Itoh Y, Elboray EE, Tsumoto H, Li Y, Suzuki M, et al. Identification of diketopiperazine-containing 2-anilinobenzamides as potent sirtuin 2 (SIRT2)-selective inhibitors targeting the selectivity pocket, substrate-binding site, and NAD+-binding site. J Med Chem 2019, 62: 5844–5862.PubMedCrossRef Mellini P, Itoh Y, Elboray EE, Tsumoto H, Li Y, Suzuki M, et al. Identification of diketopiperazine-containing 2-anilinobenzamides as potent sirtuin 2 (SIRT2)-selective inhibitors targeting the selectivity pocket, substrate-binding site, and NAD+-binding site. J Med Chem 2019, 62: 5844–5862.PubMedCrossRef
159.
Zurück zum Zitat Puentes LN, Lengyel-Zhand Z, Reilly SW, Mach RH. Evaluation of a low-toxicity PARP inhibitor as a neuroprotective agent for Parkinson’s disease. Mol Neurobiol 2021, 58: 3641–3652.PubMedCrossRef Puentes LN, Lengyel-Zhand Z, Reilly SW, Mach RH. Evaluation of a low-toxicity PARP inhibitor as a neuroprotective agent for Parkinson’s disease. Mol Neurobiol 2021, 58: 3641–3652.PubMedCrossRef
160.
Zurück zum Zitat Hamby AM, Suh SW, Kauppinen TM, Swanson RA. Use of a poly(ADP-ribose) polymerase inhibitor to suppress inflammation and neuronal death after cerebral ischemia-reperfusion. Stroke 2007, 38: 632–636.PubMedCrossRef Hamby AM, Suh SW, Kauppinen TM, Swanson RA. Use of a poly(ADP-ribose) polymerase inhibitor to suppress inflammation and neuronal death after cerebral ischemia-reperfusion. Stroke 2007, 38: 632–636.PubMedCrossRef
161.
Zurück zum Zitat Sarkar A, Dutta S, Sur M, Chakraborty S, Dey P, Mukherjee P. Early loss of endogenous NAD+ following rotenone treatment leads to mitochondrial dysfunction and Sarm1 induction that is ameliorated by PARP inhibition. FEBS J 2023, 290: 1596–1624.PubMedCrossRef Sarkar A, Dutta S, Sur M, Chakraborty S, Dey P, Mukherjee P. Early loss of endogenous NAD+ following rotenone treatment leads to mitochondrial dysfunction and Sarm1 induction that is ameliorated by PARP inhibition. FEBS J 2023, 290: 1596–1624.PubMedCrossRef
162.
Zurück zum Zitat Maier C, Scheuerle A, Hauser B, Schelzig H, Szabó C, Radermacher P, et al. The selective poly(ADP)ribose-polymerase 1 inhibitor INO1001 reduces spinal cord injury during porcine aortic cross-clamping-induced ischemia/reperfusion injury. Intensive Care Med 2007, 33: 845–850.PubMedCrossRef Maier C, Scheuerle A, Hauser B, Schelzig H, Szabó C, Radermacher P, et al. The selective poly(ADP)ribose-polymerase 1 inhibitor INO1001 reduces spinal cord injury during porcine aortic cross-clamping-induced ischemia/reperfusion injury. Intensive Care Med 2007, 33: 845–850.PubMedCrossRef
163.
Zurück zum Zitat d’Avila JC, Lam TI, Bingham D, Shi J, Won SJ, Kauppinen TM, et al. Microglial activation induced by brain trauma is suppressed by post-injury treatment with a PARP inhibitor. J Neuroinflammation 2012, 9: 31.PubMedPubMedCentralCrossRef d’Avila JC, Lam TI, Bingham D, Shi J, Won SJ, Kauppinen TM, et al. Microglial activation induced by brain trauma is suppressed by post-injury treatment with a PARP inhibitor. J Neuroinflammation 2012, 9: 31.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Cardinale A, Paldino E, Giampà C, Bernardi G, Fusco FR. PARP-1 inhibition is neuroprotective in the R6/2 mouse model of Huntington’s disease. PLoS One 2015, 10: e0134482.PubMedPubMedCentralCrossRef Cardinale A, Paldino E, Giampà C, Bernardi G, Fusco FR. PARP-1 inhibition is neuroprotective in the R6/2 mouse model of Huntington’s disease. PLoS One 2015, 10: e0134482.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Wang S, Zhang Y, Lou J, Yong H, Shan S, Liu Z, et al. The therapeutic potential of berberine chloride against SARM1-dependent axon degeneration in acrylamide-induced neuropathy. Phytother Res 2023, 37: 77–88.PubMedCrossRef Wang S, Zhang Y, Lou J, Yong H, Shan S, Liu Z, et al. The therapeutic potential of berberine chloride against SARM1-dependent axon degeneration in acrylamide-induced neuropathy. Phytother Res 2023, 37: 77–88.PubMedCrossRef
166.
Zurück zum Zitat Hughes RO, Bosanac T, Mao X, Engber TM, DiAntonio A, Milbrandt J, et al. Small molecule SARM1 inhibitors recapitulate the SARM1-/- phenotype and allow recovery of a metastable pool of axons fated to degenerate. Cell Rep 2021, 34: 108588.PubMedPubMedCentralCrossRef Hughes RO, Bosanac T, Mao X, Engber TM, DiAntonio A, Milbrandt J, et al. Small molecule SARM1 inhibitors recapitulate the SARM1-/- phenotype and allow recovery of a metastable pool of axons fated to degenerate. Cell Rep 2021, 34: 108588.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Bratkowski M, Burdett TC, Danao J, Wang X, Mathur P, Gu W, et al. Uncompetitive, adduct-forming SARM1 inhibitors are neuroprotective in preclinical models of nerve injury and disease. Neuron 2022, 110: 3711–3726.e16.PubMedCrossRef Bratkowski M, Burdett TC, Danao J, Wang X, Mathur P, Gu W, et al. Uncompetitive, adduct-forming SARM1 inhibitors are neuroprotective in preclinical models of nerve injury and disease. Neuron 2022, 110: 3711–3726.e16.PubMedCrossRef
168.
Zurück zum Zitat Feldman HC, Merlini E, Guijas C, DeMeester KE, Njomen E, Kozina EM, et al. Selective inhibitors of SARM1 targeting an allosteric cysteine in the autoregulatory ARM domain. Proc Natl Acad Sci U S A 2022, 119: e2208457119.PubMedPubMedCentralCrossRef Feldman HC, Merlini E, Guijas C, DeMeester KE, Njomen E, Kozina EM, et al. Selective inhibitors of SARM1 targeting an allosteric cysteine in the autoregulatory ARM domain. Proc Natl Acad Sci U S A 2022, 119: e2208457119.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Tarragó MG, Chini CCS, Kanamori KS, Warner GM, Caride A, de Oliveira GC, et al. A potent and specific CD38 inhibitor ameliorates age-related metabolic dysfunction by reversing tissue NAD+ decline. Cell Metab 2018, 27: 1081–1095.e10.PubMedPubMedCentralCrossRef Tarragó MG, Chini CCS, Kanamori KS, Warner GM, Caride A, de Oliveira GC, et al. A potent and specific CD38 inhibitor ameliorates age-related metabolic dysfunction by reversing tissue NAD+ decline. Cell Metab 2018, 27: 1081–1095.e10.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Peclat TR, Thompson KL, Warner GM, Chini CCS, Tarragó MG, Mazdeh DZ, et al. CD38 inhibitor 78c increases mice lifespan and healthspan in a model of chronological aging. Aging Cell 2022, 21: e13589.PubMedPubMedCentralCrossRef Peclat TR, Thompson KL, Warner GM, Chini CCS, Tarragó MG, Mazdeh DZ, et al. CD38 inhibitor 78c increases mice lifespan and healthspan in a model of chronological aging. Aging Cell 2022, 21: e13589.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Roboon J, Hattori T, Ishii H, Takarada-Iemata M, Nguyen DT, Heer CD, et al. Inhibition of CD38 and supplementation of nicotinamide riboside ameliorate lipopolysaccharide-induced microglial and astrocytic neuroinflammation by increasing NAD. J Neurochem 2021, 158: 311–327.PubMedPubMedCentralCrossRef Roboon J, Hattori T, Ishii H, Takarada-Iemata M, Nguyen DT, Heer CD, et al. Inhibition of CD38 and supplementation of nicotinamide riboside ameliorate lipopolysaccharide-induced microglial and astrocytic neuroinflammation by increasing NAD. J Neurochem 2021, 158: 311–327.PubMedPubMedCentralCrossRef
172.
Zurück zum Zitat Ogura Y, Kitada M, Xu J, Monno I, Koya D. CD38 inhibition by apigenin ameliorates mitochondrial oxidative stress through restoration of the intracellular NAD+/NADH ratio and Sirt3 activity in renal tubular cells in diabetic rats. Aging (Albany NY) 2020, 12: 11325–11336.PubMedCrossRef Ogura Y, Kitada M, Xu J, Monno I, Koya D. CD38 inhibition by apigenin ameliorates mitochondrial oxidative stress through restoration of the intracellular NAD+/NADH ratio and Sirt3 activity in renal tubular cells in diabetic rats. Aging (Albany NY) 2020, 12: 11325–11336.PubMedCrossRef
173.
Zurück zum Zitat Li BS, Zhu RZ, Lim SH, Seo JH, Choi BM. Apigenin Alleviates Oxidative Stress-Induced Cellular Senescence via Modulation of the SIRT1-NAD[Formula: See text]-CD38 Axis. Am J Chin Med 2021, 49: 1235–1250.PubMedCrossRef Li BS, Zhu RZ, Lim SH, Seo JH, Choi BM. Apigenin Alleviates Oxidative Stress-Induced Cellular Senescence via Modulation of the SIRT1-NAD[Formula: See text]-CD38 Axis. Am J Chin Med 2021, 49: 1235–1250.PubMedCrossRef
174.
Zurück zum Zitat Lagu B, Wu X, Kulkarni S, Paul R, Wu X, Olson L, et al. Orally bioavailable enzymatic inhibitor of CD38, MK-0159, protects against ischemia/reperfusion injury in the murine heart. J Med Chem 2022, 65: 9418–9446.PubMedCrossRef Lagu B, Wu X, Kulkarni S, Paul R, Wu X, Olson L, et al. Orally bioavailable enzymatic inhibitor of CD38, MK-0159, protects against ischemia/reperfusion injury in the murine heart. J Med Chem 2022, 65: 9418–9446.PubMedCrossRef
175.
Zurück zum Zitat Bhat A, Tan V, Heng B, Chow S, Basappa S, Essa MM, et al. Papaverine, a phosphodiesterase 10A inhibitor, ameliorates quinolinic acid-induced synaptotoxicity in human cortical neurons. Neurotox Res 2021, 39: 1238–1250.PubMedCrossRef Bhat A, Tan V, Heng B, Chow S, Basappa S, Essa MM, et al. Papaverine, a phosphodiesterase 10A inhibitor, ameliorates quinolinic acid-induced synaptotoxicity in human cortical neurons. Neurotox Res 2021, 39: 1238–1250.PubMedCrossRef
176.
Zurück zum Zitat Bhat A, Tan V, Heng B, Lovejoy DB, Sakharkar MK, Essa MM, et al. Roflumilast, a cAMP-specific phosphodiesterase-4 inhibitor, reduces oxidative stress and improves synapse functions in human cortical neurons exposed to the excitotoxin quinolinic acid. ACS Chem Neurosci 2020, 11: 4405–4415.PubMedCrossRef Bhat A, Tan V, Heng B, Lovejoy DB, Sakharkar MK, Essa MM, et al. Roflumilast, a cAMP-specific phosphodiesterase-4 inhibitor, reduces oxidative stress and improves synapse functions in human cortical neurons exposed to the excitotoxin quinolinic acid. ACS Chem Neurosci 2020, 11: 4405–4415.PubMedCrossRef
177.
Zurück zum Zitat Zhong J, Dong W, Qin Y, Xie J, Xiao J, Xu J, et al. Roflupram exerts neuroprotection via activation of CREB/PGC-1α signalling in experimental models of Parkinson’s disease. Br J Pharmacol 2020, 177: 2333–2350.PubMedPubMedCentralCrossRef Zhong J, Dong W, Qin Y, Xie J, Xiao J, Xu J, et al. Roflupram exerts neuroprotection via activation of CREB/PGC-1α signalling in experimental models of Parkinson’s disease. Br J Pharmacol 2020, 177: 2333–2350.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Dong WL, Zhong JH, Chen YQ, Xie JF, Qin YY, Xu JP, et al. Roflupram protects against rotenone-induced neurotoxicity and facilitates α-synuclein degradation in Parkinson’s disease models. Acta Pharmacol Sin 2021, 42: 1991–2003.PubMedPubMedCentralCrossRef Dong WL, Zhong JH, Chen YQ, Xie JF, Qin YY, Xu JP, et al. Roflupram protects against rotenone-induced neurotoxicity and facilitates α-synuclein degradation in Parkinson’s disease models. Acta Pharmacol Sin 2021, 42: 1991–2003.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: Promises, achievements, and challenges. Pharmacol Ther 2018, 188: 140–154.PubMedPubMedCentralCrossRef Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: Promises, achievements, and challenges. Pharmacol Ther 2018, 188: 140–154.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Bonkowski MS, Sinclair DA. Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol 2016, 17: 679–690.PubMedPubMedCentralCrossRef Bonkowski MS, Sinclair DA. Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds. Nat Rev Mol Cell Biol 2016, 17: 679–690.PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Campagna J, Spilman P, Jagodzinska B, Bai D, Hatami A, Zhu C, et al. A small molecule ApoE4-targeted therapeutic candidate that normalizes sirtuin 1 levels and improves cognition in an Alzheimer’s disease mouse model. Sci Rep 2018, 8: 17574.ADSPubMedPubMedCentralCrossRef Campagna J, Spilman P, Jagodzinska B, Bai D, Hatami A, Zhu C, et al. A small molecule ApoE4-targeted therapeutic candidate that normalizes sirtuin 1 levels and improves cognition in an Alzheimer’s disease mouse model. Sci Rep 2018, 8: 17574.ADSPubMedPubMedCentralCrossRef
183.
Zurück zum Zitat Lu Y, Tan L, Wang X. Circular HDAC9/microRNA-138/sirtuin-1 pathway mediates synaptic and amyloid precursor protein processing deficits in alzheimer’s disease. Neurosci Bull 2019, 35: 877–888.PubMedPubMedCentralCrossRef Lu Y, Tan L, Wang X. Circular HDAC9/microRNA-138/sirtuin-1 pathway mediates synaptic and amyloid precursor protein processing deficits in alzheimer’s disease. Neurosci Bull 2019, 35: 877–888.PubMedPubMedCentralCrossRef
184.
Zurück zum Zitat Ross AG, Chaqour B, McDougald DS, Dine KE, Duong TT, Shindler RE, et al. Selective upregulation of SIRT1 expression in retinal ganglion cells by AAV-mediated gene delivery increases neuronal cell survival and alleviates axon demyelination associated with optic neuritis. Biomolecules 2022, 12: 830.PubMedPubMedCentralCrossRef Ross AG, Chaqour B, McDougald DS, Dine KE, Duong TT, Shindler RE, et al. Selective upregulation of SIRT1 expression in retinal ganglion cells by AAV-mediated gene delivery increases neuronal cell survival and alleviates axon demyelination associated with optic neuritis. Biomolecules 2022, 12: 830.PubMedPubMedCentralCrossRef
185.
Zurück zum Zitat Chandrasekaran K, Salimian M, Konduru SR, Choi J, Kumar P, Long A, et al. Overexpression of Sirtuin 1 protein in neurons prevents and reverses experimental diabetic neuropathy. Brain 2019, 142: 3737–3752.PubMedPubMedCentralCrossRef Chandrasekaran K, Salimian M, Konduru SR, Choi J, Kumar P, Long A, et al. Overexpression of Sirtuin 1 protein in neurons prevents and reverses experimental diabetic neuropathy. Brain 2019, 142: 3737–3752.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Verma R, Ritzel RM, Crapser J, Friedler BD, McCullough LD. Evaluation of the neuroprotective effect of Sirt3 in experimental stroke. Transl Stroke Res 2019, 10: 57–66.PubMedCrossRef Verma R, Ritzel RM, Crapser J, Friedler BD, McCullough LD. Evaluation of the neuroprotective effect of Sirt3 in experimental stroke. Transl Stroke Res 2019, 10: 57–66.PubMedCrossRef
187.
Zurück zum Zitat Wang Y, Yang J, Hong T, Chen X, Cui L. SIRT2: Controversy and multiple roles in disease and physiology. Ageing Res Rev 2019, 55: 100961.PubMedCrossRef Wang Y, Yang J, Hong T, Chen X, Cui L. SIRT2: Controversy and multiple roles in disease and physiology. Ageing Res Rev 2019, 55: 100961.PubMedCrossRef
188.
Zurück zum Zitat Stein LR, Imai SI. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J 2014, 33: 1321–1340.PubMedPubMedCentral Stein LR, Imai SI. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J 2014, 33: 1321–1340.PubMedPubMedCentral
189.
Zurück zum Zitat Luo H, Mu WC, Karki R, Chiang HH, Mohrin M, Shin JJ, et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Rep 2019, 26: 945-954.e4.PubMedPubMedCentralCrossRef Luo H, Mu WC, Karki R, Chiang HH, Mohrin M, Shin JJ, et al. Mitochondrial stress-initiated aberrant activation of the NLRP3 inflammasome regulates the functional deterioration of hematopoietic stem cell aging. Cell Rep 2019, 26: 945-954.e4.PubMedPubMedCentralCrossRef
190.
Zurück zum Zitat Ma XR, Zhu X, Xiao Y, Gu HM, Zheng SS, Li L, et al. Restoring nuclear entry of Sirtuin 2 in oligodendrocyte progenitor cells promotes remyelination during ageing. Nat Commun 2022, 13: 1225.ADSPubMedPubMedCentralCrossRef Ma XR, Zhu X, Xiao Y, Gu HM, Zheng SS, Li L, et al. Restoring nuclear entry of Sirtuin 2 in oligodendrocyte progenitor cells promotes remyelination during ageing. Nat Commun 2022, 13: 1225.ADSPubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Chamberlain KA, Huang N, Xie Y, Li Causi F, Li S, Li Y, et al. Oligodendrocytes enhance axonal energy metabolism by deacetylation of mitochondrial proteins through transcellular delivery of SIRT2. Neuron 2021, 109: 3456-3472.e8.PubMedPubMedCentralCrossRef Chamberlain KA, Huang N, Xie Y, Li Causi F, Li S, Li Y, et al. Oligodendrocytes enhance axonal energy metabolism by deacetylation of mitochondrial proteins through transcellular delivery of SIRT2. Neuron 2021, 109: 3456-3472.e8.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat She DT, Wong LJ, Baik SH, Arumugam TV. SIRT2 inhibition confers neuroprotection by downregulation of FOXO3a and MAPK signaling pathways in ischemic stroke. Mol Neurobiol 2018, 55: 9188–9203.PubMedCrossRef She DT, Wong LJ, Baik SH, Arumugam TV. SIRT2 inhibition confers neuroprotection by downregulation of FOXO3a and MAPK signaling pathways in ischemic stroke. Mol Neurobiol 2018, 55: 9188–9203.PubMedCrossRef
193.
Zurück zum Zitat Narayan N, Lee IH, Borenstein R, Sun J, Wong R, Tong G, et al. The NAD-dependent deacetylase SIRT2 is required for programmed necrosis. Nature 2012, 492: 199–204.ADSPubMedCrossRef Narayan N, Lee IH, Borenstein R, Sun J, Wong R, Tong G, et al. The NAD-dependent deacetylase SIRT2 is required for programmed necrosis. Nature 2012, 492: 199–204.ADSPubMedCrossRef
194.
Zurück zum Zitat D’Andrea AD. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair 2018, 71: 172–176.PubMedCrossRef D’Andrea AD. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair 2018, 71: 172–176.PubMedCrossRef
195.
Zurück zum Zitat Teng F, Zhu L, Su J, Zhang X, Li N, Nie Z, et al. Neuroprotective effects of poly(ADP-ribose)polymerase inhibitor olaparib in transient cerebral ischemia. Neurochem Res 2016, 41: 1516–1526.PubMedCrossRef Teng F, Zhu L, Su J, Zhang X, Li N, Nie Z, et al. Neuroprotective effects of poly(ADP-ribose)polymerase inhibitor olaparib in transient cerebral ischemia. Neurochem Res 2016, 41: 1516–1526.PubMedCrossRef
196.
Zurück zum Zitat Virág L, Szabó C. Purines inhibit poly(ADP-ribose) polymerase activation and modulate oxidant-induced cell death. FASEB J 2001, 15: 99–107.PubMedCrossRef Virág L, Szabó C. Purines inhibit poly(ADP-ribose) polymerase activation and modulate oxidant-induced cell death. FASEB J 2001, 15: 99–107.PubMedCrossRef
197.
Zurück zum Zitat Shi Y, Kerry PS, Nanson JD, Bosanac T, Sasaki Y, Krauss R, et al. Structural basis of SARM1 activation, substrate recognition, and inhibition by small molecules. Mol Cell 2022, 82: 1643-1659.e10.PubMedPubMedCentralCrossRef Shi Y, Kerry PS, Nanson JD, Bosanac T, Sasaki Y, Krauss R, et al. Structural basis of SARM1 activation, substrate recognition, and inhibition by small molecules. Mol Cell 2022, 82: 1643-1659.e10.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Henninger N, Bouley J, Sikoglu EM, An J, Moore CM, King JA, et al. Attenuated traumatic axonal injury and improved functional outcome after traumatic brain injury in mice lacking Sarm1. Brain 2016, 139: 1094–1105.PubMedPubMedCentralCrossRef Henninger N, Bouley J, Sikoglu EM, An J, Moore CM, King JA, et al. Attenuated traumatic axonal injury and improved functional outcome after traumatic brain injury in mice lacking Sarm1. Brain 2016, 139: 1094–1105.PubMedPubMedCentralCrossRef
199.
Zurück zum Zitat Geisler S, Doan RA, Strickland A, Huang X, Milbrandt J, DiAntonio A. Prevention of vincristine-induced peripheral neuropathy by genetic deletion of SARM1 in mice. Brain 2016, 139: 3092–3108.PubMedPubMedCentralCrossRef Geisler S, Doan RA, Strickland A, Huang X, Milbrandt J, DiAntonio A. Prevention of vincristine-induced peripheral neuropathy by genetic deletion of SARM1 in mice. Brain 2016, 139: 3092–3108.PubMedPubMedCentralCrossRef
200.
201.
Zurück zum Zitat Sasaki Y, Zhu J, Shi Y, Gu W, Kobe B, Ve T, et al. Nicotinic acid mononucleotide is an allosteric SARM1 inhibitor promoting axonal protection. Exp Neurol 2021, 345: 113842.PubMedPubMedCentralCrossRef Sasaki Y, Zhu J, Shi Y, Gu W, Kobe B, Ve T, et al. Nicotinic acid mononucleotide is an allosteric SARM1 inhibitor promoting axonal protection. Exp Neurol 2021, 345: 113842.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Li Y, Yang R, Chen L, Wu S. CD38 as an immunomodulator in cancer. Future Oncol 2020, 16: 2853–2861.PubMedCrossRef Li Y, Yang R, Chen L, Wu S. CD38 as an immunomodulator in cancer. Future Oncol 2020, 16: 2853–2861.PubMedCrossRef
203.
Zurück zum Zitat Hogan KA, Chini CCS, Chini EN. The multi-faceted ecto-enzyme CD38: Roles in immunomodulation, cancer, aging, and metabolic diseases. Front Immunol 2019, 10: 1187.PubMedPubMedCentralCrossRef Hogan KA, Chini CCS, Chini EN. The multi-faceted ecto-enzyme CD38: Roles in immunomodulation, cancer, aging, and metabolic diseases. Front Immunol 2019, 10: 1187.PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Blacher E, Dadali T, Bespalko A, Haupenthal VJ, Grimm MOW, Hartmann T, et al. Alzheimer’s disease pathology is attenuated in a CD38-deficient mouse model. Ann Neurol 2015, 78: 88–103.PubMedPubMedCentralCrossRef Blacher E, Dadali T, Bespalko A, Haupenthal VJ, Grimm MOW, Hartmann T, et al. Alzheimer’s disease pathology is attenuated in a CD38-deficient mouse model. Ann Neurol 2015, 78: 88–103.PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Long A, Park JH, Klimova N, Fowler C, Loane DJ, Kristian T. CD38 knockout mice show significant protection against ischemic brain damage despite high level poly-ADP-ribosylation. Neurochem Res 2017, 42: 283–293.PubMedCrossRef Long A, Park JH, Klimova N, Fowler C, Loane DJ, Kristian T. CD38 knockout mice show significant protection against ischemic brain damage despite high level poly-ADP-ribosylation. Neurochem Res 2017, 42: 283–293.PubMedCrossRef
206.
Zurück zum Zitat Neelakantan H, Brightwell CR, Graber TG, Maroto R, Wang HYL, McHardy SF, et al. Small molecule nicotinamide N-methyltransferase inhibitor activates senescent muscle stem cells and improves regenerative capacity of aged skeletal muscle. Biochem Pharmacol 2019, 163: 481–492.PubMedPubMedCentralCrossRef Neelakantan H, Brightwell CR, Graber TG, Maroto R, Wang HYL, McHardy SF, et al. Small molecule nicotinamide N-methyltransferase inhibitor activates senescent muscle stem cells and improves regenerative capacity of aged skeletal muscle. Biochem Pharmacol 2019, 163: 481–492.PubMedPubMedCentralCrossRef
207.
Zurück zum Zitat Sonavane M, Hayat F, Makarov M, Migaud ME, Gassman NR. Dihydronicotinamide riboside promotes cell-specific cytotoxicity by tipping the balance between metabolic regulation and oxidative stress. PLoS One 2020, 15: e0242174.PubMedPubMedCentralCrossRef Sonavane M, Hayat F, Makarov M, Migaud ME, Gassman NR. Dihydronicotinamide riboside promotes cell-specific cytotoxicity by tipping the balance between metabolic regulation and oxidative stress. PLoS One 2020, 15: e0242174.PubMedPubMedCentralCrossRef
Metadaten
Titel
Targeting NAD Metabolism for the Therapy of Age-Related Neurodegenerative Diseases
verfasst von
Feifei Li
Chou Wu
Gelin Wang
Publikationsdatum
31.05.2023
Verlag
Springer Nature Singapore
Erschienen in
Neuroscience Bulletin / Ausgabe 2/2024
Print ISSN: 1673-7067
Elektronische ISSN: 1995-8218
DOI
https://doi.org/10.1007/s12264-023-01072-3

Weitere Artikel der Ausgabe 2/2024

Neuroscience Bulletin 2/2024 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Was nützt die Kraniektomie bei schwerer tiefer Hirnblutung?

17.05.2024 Hirnblutung Nachrichten

Eine Studie zum Nutzen der druckentlastenden Kraniektomie nach schwerer tiefer supratentorieller Hirnblutung deutet einen Nutzen der Operation an. Für überlebende Patienten ist das dennoch nur eine bedingt gute Nachricht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.